EP1335745A1 - Use of a compound antagonist of the esm-1 protein for producing a medicine for treating cancer - Google Patents

Use of a compound antagonist of the esm-1 protein for producing a medicine for treating cancer

Info

Publication number
EP1335745A1
EP1335745A1 EP01993475A EP01993475A EP1335745A1 EP 1335745 A1 EP1335745 A1 EP 1335745A1 EP 01993475 A EP01993475 A EP 01993475A EP 01993475 A EP01993475 A EP 01993475A EP 1335745 A1 EP1335745 A1 EP 1335745A1
Authority
EP
European Patent Office
Prior art keywords
esm
protein
compound
antagonist
antagonist compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP01993475A
Other languages
German (de)
French (fr)
Inventor
Philippe Résidence Sébastopol LASSALLE
David Bechard
André-Bernard TONNEL
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institut Pasteur de Lille
Institut National de la Sante et de la Recherche Medicale INSERM
Original Assignee
Institut Pasteur de Lille
Institut National de la Sante et de la Recherche Medicale INSERM
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut Pasteur de Lille, Institut National de la Sante et de la Recherche Medicale INSERM filed Critical Institut Pasteur de Lille
Publication of EP1335745A1 publication Critical patent/EP1335745A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to the field of cancer prevention and / or treatment.
  • Cancer is frequently a disease linked to deficits in the intracellular signaling system. Normal cells respond to many extracellular signals by proliferating, differentiating or more generally by altering their metabolic activity.
  • Such signals are received on the surface of cells and converted by a signal transduction protein system into a message recognized by the cell. This message is responsible for phenomena of subsequent cellular regulation.
  • Metastasis is the formation of a tumor colony
  • Tumor cells locally escape through tissue barriers, such as the basement membrane of the epithelium, and reach the interstitial stroma, from which they access blood vessels
  • the circulating tumor cells are carried into the bloodstream and are stopped in the precapillary venules of the target organ by adhesion to the lumen surfaces of the endothelial cell, or are
  • the tumor cells leave the vascular wall and enter the parenchyma of the organ. Finally, the tumor cell, after extravasation, multiplies in a tissue different from that from which it originates.
  • a number of cancers have been shown to be caused by defects associated with genes responsible for signal transduction. Such genes are called encogenes. Oncogenes can lead to overexpression of one or more signal transducing proteins inducing abnormal cell proliferation. Defective signals can be linked to various mechanisms. Certain anticancer therapies aim to inhibit the expression or the bioavailability of oncogenic proteins responsible for the proliferation of cancer cells, such as proteins of the MAP kinase family or products of certain oncogenes such as c-myc.
  • the ESM-1 protein is a polypeptide of 184 amino acids secreted by endothelial cells and which was described for the first time by LASSALLE et al. (1996).
  • the messenger RNAs coding for the protein ESM-1 are mainly found in endothelial cells and in pulmonary and renal tissues.
  • the expression of the gene encoding ESM-1 is regulated by cytokines. TNF- ⁇ and IL-1 ⁇ induce an increase in the expression of the ESM-1 gene in endothelial cells of the human umbilical vein, while Interferon- ⁇ decreases its expression.
  • ESM-1 protein A large amount of circulating ESM-1 protein has been found in patients with systemic inflammatory syndrome, such as septic shock (BECHARD et al., (2000).
  • the object of the present invention is to provide anti-cancer compounds which would make it possible to overcome the disadvantages of State of the art methods of therapeutic treatment of cancer.
  • a first object of the invention consists in the use of a compound antagonist of the ESM-1 protein for the manufacture of a medicament for the treatment of cancer.
  • an antagonist compound of the invention is an antibody specifically binding to the protein ESM-1.
  • an antagonist compound used in the context of the invention is a peptide of at least 10 amino acids of the modified ESM-1 protein and which comprises the chain of amino acids Ala (134) - Ala (135).
  • an antagonist compound of the ESM-1 protein consists of an antisense oligonucleotide hybridizing with the cDNA coding for ESM-1.
  • Another object of the invention consists of an antagonist compound of the ESM-1 protein, chosen from the antagonist compounds defined above.
  • the invention also relates to a pharmaceutical composition intended for the treatment of cancer comprising an antagonist compound of the protein ESM-1.
  • Another subject of the invention consists of a method of preventing cancer comprising a step during which a compound antagonist of the protein ESM-1 is administered.
  • the invention also relates to a method of therapeutic treatment of cancer comprising a step during which a compound antagonist of the protein ESM-1 is administered to a patient.
  • the protein ESM-1 is secreted in humans in the form of a proteoglycan of the chondroitin / dermatan sulfate type and that the protein Secreted ESM-1 is capable of stimulating the mitogenic activity of factor HGF / SF in vitro (for “Hepatocyte growth factor / scatter factor”).
  • HGF / SF is an important factor in the onset of renal multicystic dysplasia and in the appearance of hyperproliferation of the renal tubules and has also been associated with the development of carcinomas of the breast, kidneys and lung but also in the development of malignant melanomas.
  • transfected human renal epithelial cells expressing the protein ESM-1 have a high tumor potential and cause the appearance of renal carcinoma in vivo in mice.
  • Antibodies against the ESM-1 protein have also been shown to be capable of inhibiting the development of a renal tumor in vivo and that an antagonist peptide of the ESM-1 protein has the same anti-tumor activity.
  • an increase in the serum ESM-1 protein level has been shown according to the invention in patients with bronchopulmonary carcinoma.
  • a first object of the invention consists in the use of an antagonist compound of the ESM-1 protein for the manufacture of a medicament for the prevention and / or treatment of cancer.
  • ESM-1 protein or “ESM-1 polypeptide”, within the meaning of the invention, include a polypeptide of 184 amino acids referenced as the sequence SEQ ID No. 1 of the sequence listing, as well as a polypeptide of 165 amino acids identical to the polypeptide of sequence SEQ ID No. 1 in which the 19 amino acids of the N-terminal corresponding to the signal peptides are absent, this polypeptide of 165 amino acids constituting the secreted form of the polypeptide of sequence SEQ ID N 1. Also included in the definition of “ESM-1 protein” “ESM-1 polypeptide” is a glycopeptide of 184 amino acids of sequence SEQ ID No.
  • the ESM-1 glycopeptide has the serine residue at position 137 which is O-glycosylated by a chondroitin / dermatan sulfate motif.
  • an antagonist compound of the protein ESM-1 is meant according to the invention a compound capable of significantly reducing the bioavailability of the protein ESM-1 with respect to target molecules on which which the protein ESM-1 is naturally fixes.
  • An ESM-1 protein antagonist compound can reduce the bioavailability of these proteins by reducing the likelihood of binding of the ESM-1 protein to target molecules in the body to which it naturally binds.
  • An antagonist compound according to the invention can reduce the bioavailability of the protein ESM-1 by inhibiting or blocking the transcription of the gene coding for ESM-1, by inhibiting or blocking the translation of the corresponding messenger RNA, by altering the intracellular processing of the ESM-1 protein, for example by affecting the enzymatic process leading to its glycosylation, or by inhibiting or blocking the secretion of the mature ESM-1 protein.
  • a first object of the invention consists in the use of a compound antagonist of the ESM-1 protein for the manufacture of a medicament for the treatment of cancer.
  • An ESM-1 protein antagonist compound may be of any kind, polypeptide, saccharide, or any organic or inorganic compound making it possible to reduce the bioavailability of the ESM-1 protein with respect to the target molecules on which this protein is fixed.
  • a first family of ESM-1 antagonist compounds preferred according to the invention consists of antibodies specifically binding to the ESM-1 protein. It is shown according to the invention that antibodies directed specifically against the protein ESM-1 are capable of inhibiting or blocking the tumorigenicity of this protein. Anti-ESM-1 antibodies therefore constitute antagonistic compounds of great therapeutic value.
  • antibody within the meaning of the invention is meant in particular polyclonal or monoclonal antibodies or fragments (for example the Fab or F (ab) ' 2 fragments or any polypeptide comprising a domain of the initial antibody recognizing the protein ESM-1.
  • Monoclonal antibodies can be prepared from a hybridoma using the technique described by KOHLER and MIELSTEIN (1975).
  • ESM-1 or a fragment of this protein produced by the trioma technique or also the hybridoma technique described by KOZBOR et al. (1983).
  • Anti-ESM-1 antibodies according to the invention can also be fragments of single chain Fv antibody (ScFv) such as those described in US Patent No. 0 4,9476,778 or by MARTINEAU et al. (1998).
  • Anti-ESM-1 antibodies according to the invention also comprise fragments of antibodies obtained using phage banks as described by RIDDER et al. (1995) or humanized antibodies as described by REINMANN et al. (1997) or also by LEGER OJ, et al., 1997. They may also be anti-ESM-1 antibodies produced according to the techniques described by BECHARD et al. (2000). The antibodies described by BECHARD et al.
  • a first preferred family of antibodies according to the invention which constitute antagonist compounds of the ESM-1 protein are the monoclonal antibodies specifically recognizing the region going from the praline residue in position 79 to the cysteine residue in position 99 of the sequence SEQ ID No. 1, this region representing the antigenic determinant D1. They are preferably monoclonal antibodies produced by the hybridoma line deposited on November 19, 1997 with the National Collection of Cultures of Microorganisms of the Pasteur Institute (CNCM) under the access number N ° l -1944, also called MEP21 antibody.
  • CNCM National Collection of Cultures of Microorganisms of the Pasteur Institute
  • preferred monoclonal antibodies are they which specifically bind to the part of the ESM-1 protein between the glycine residue at position 159 and the arginine residue at position 184 of the sequence SEQ ID No. 1 which is the region constituting the determinant D3 antigen.
  • Preferred monoclonal antibodies specific for the antigenic determinant D3 can be obtained from the hybridoma line 1-1943 (MEP19), deposited on November 19, 1997 with the National Collection of Cultures of Microorganisms of the Institut Pasteur (CNCM ).
  • Other preferred monoclonal antibodies according to the invention are the monoclonal antibodies binding specifically with the region between the serine residue at position 119 and the valine residue at position 139 of the protein ESM-1 of sequence SEQ ID No.
  • the antigenic determinant D2 of the protein ESM-1 is defined as the antigenic determinant D2 of the protein ESM-1.
  • the preferred monoclonal antibodies specifically binding to the antigenic determinant D2 of ESM-1 can be obtained from the hybridoma line MEP08 deposited on November 19, 1997 with the National Collection of Cultures of Microorganisms of the Institut Pasteur (CNCM) under the access number N ° 1-1941.
  • Other monoclonal antibodies of interest constituting antagonist compounds of the ESM-1 protein are the monoclonal antibodies directed specifically against the N-terminal part of the ESM-1 protein.
  • the preferred monoclonal antibodies directed against the N-terminal part of the ESM-1 protein can be obtained from the hybridoma line MEC15 deposited in the Collection National Culture of Microorganisms of the INSTITUT PASTEUR (CNCM) on October 17, 2000 under the access number I-2572.
  • the anti-ESM1 antibodies having the best antagonistic activities vis-à-vis ESM-1 are chosen from antibodies specifically recognizing epitopes located in the region surrounding the phenylalanine residue at position 115. It s acts in particular antibodies which bind specifically with the region between the serine residue at position 119 and the valine residue at position 139 of the protein ESM-1 of sequence SEQ ID No. 1, such as the monoclonal antibody MEP08 described above above.
  • the monoclonal antibody MEP08 was capable of inhibiting the pro-tumor activity of the protein ESM-1 on the formation of tumors caused by the proliferation of human cells of renal origin in mice .
  • the region covering the antigenic determinant D2 of the protein ESM-1 is important for the pro-tumor activity of the protein ESM-1.
  • the applicant has synthesized a polypeptide derived from the ESM-1 protein in which the phenylalanine residues in positions 134 and 135 of the sequence SEQ ID No. 1, that is to say the residues in positions 115 and 116 of the secreted protein ESM-1, have been replaced by two alanine residues.
  • This modified polypeptide is incapable of inducing tumors in mice.
  • Such a modified polypeptide can therefore compete with the protein ESM-1 produced at high level in cancer patients for its potentiating action with growth factors such as HGF / SF or even growth factors FGF-2 and FGF-7 .
  • ESM-1 protein antagonist compounds include polypeptides of at least 10 consecutive amino acids in sequence SEQ ID NO: 1, which include an amino acid sequence ranging from amino acid at position 119 to with the amino acid in position 139 of the sequence SEQ ID No. 1, such a polypeptide ESM-1 antagonist comprising at least one substitution of an amino acid, with respect to the corresponding sequence of the ESM-1 protein.
  • an ESM-1 protein antagonist polypeptide as defined above has at most 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 , 30, 35, 40, 45 or 50 consecutive amino acids of the sequence SEQ ID No 1 and at least one substitution of amino acids, compared to the sequence SEQ ID No 1.
  • An ESM-1 protein antagonist polypeptide comprises at most 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid substitutions, relative to the sequence SEQ ID No 1, the number of amino acid substitutions being adapted as a function of the length of the polypeptide, it being understood that the number of amino acid substitutions relative to the sequence SEQ ID No 1 in an antagonistic polypeptide according to the invention is at most 25% of the amino acids included in the sequence of this antagonistic polypeptide, preferably at most 20%, 15% and preferably at most 10% of the number of amino acids included in the ESM-1 antagonist polypeptide sequence.
  • an amino acid substitution, with respect to the sequence SEQ ID No. 1, in an antagonistic polypeptide according to the invention is a so-called “non-conservative” substitution.
  • non-conservative substitution is meant the substitution of an amino acid residue with an amino acid of a distinct class.
  • Amino acids are classically classified according to the following classes:
  • - non-polar (hydrophobic) amino acids alanine, leucine, isoleucine, valine, praline, phenylalanine, tryptophan and methionine;
  • - amino acids containing aromatic rings phenylalanine, tryptophan and tyrosine
  • - polar neutral amino acids glycine, serine, threonine, cysteine, tyrosine, asparagine and glutamine;
  • an ESM-1 protein antagonist polypeptide for the preparation of an ESM-1 protein antagonist polypeptide according to the invention is the substitution of an amino acid containing an aromatic ring with an amino acid not containing an aromatic ring.
  • an antagonist polypeptide of the ESM-1 protein according to the invention comprises a substitution of the phenylalanine residues in positions 134 and 135 of SEQ ID No. 1 with two amino acid residues, identical or different, not containing an aromatic cycle.
  • Such a preferred ESM-1 protein antagonist polypeptide is a polypeptide of at least 10 consecutive amino acids of the sequence SEQ ID No.
  • an antagonist polypeptide of the ESM-1 protein according to the invention can be prepared by conventional techniques of chemical synthesis, either in homogeneous solution or in solid phase.
  • an ESM-1 protein antagonist polypeptide can be prepared by the homogeneous solution technique described by HOUBEN WEIL (1974) or the solid phase synthesis technique described by MERRIFIELD (1965a; 1965b) and MERRIFIELD 1965b.
  • An ESM-1 protein antagonist polypeptide according to the invention can also be prepared by genetic recombination.
  • a method can be implemented comprising the steps of: a) inserting a nucleic acid coding for the ESM-1 protein antagonist polypeptide in a vector appropriate expression; b) cultivating, in an appropriate culture medium, a host cell previously transformed or transfected with the recombinant expression vector of step a); c) recovering the conditioned culture medium or lysing the host cell, for example by sonication or by osmotic shock; d) separating and purifying from said culture medium or also from the cell lysates obtained in step c), said antagonist polypeptide; e) where appropriate, characterize the recombinant antagonist polypeptide thus produced.
  • the antagonistic polypeptides according to the invention can be characterized by fixation on an immunoaffinity chromatography column on which the antibodies directed against this polypeptide or against a fragment of the latter have been immobilized beforehand.
  • an ESM-1 antagonist polypeptide can be purified by passage through an appropriate series of chromatography columns, according to methods known to those skilled in the art is described for example in AUSUBEL F. et al. (1989).
  • ESM-1 antagonist compounds aimed at reducing the bioavailability of the secreted ESM-1 protein in patients at risk or in patients who have already developed tumors are compounds capable of inhibiting or blocking expression of the gene encoding ESM-1 in humans.
  • ESM-1 protein antagonist compounds can be antisense polynucleotides.
  • the antagonist compounds of the ESM-1 protein according to the invention thus include an antisense polynucleotide capable of hybridizing specifically to a determined region of the gene coding for the protein ESM-1 and capable of inhibiting or blocking its transcription and / or its translation.
  • the sequence of the human ESM-1 gene is referenced under the access number AJ401 1091 and AJ401 1092 in the Genbank database.
  • an antisense polynucleotide according to the invention comprises a sequence complementary to a sequence localized in the region of the 5 ′ end of the DNA of the ESM-1 gene, and so completely preferred is the proximity of the translation initiation codon (ATG) of the ESM-1 gene.
  • ATG translation initiation codon
  • an antisense polynucleotide according to the invention comprises a sequence complementary to one of the sequences located at the exon / intron junctions of the ESM-1 gene and preferably sequences corresponding to a site of splicing.
  • a preferred antisense polynucleotide according to the invention comprises at least 15 consecutive nucleotides of the cDNA coding for ESM-1 having the nucleotide sequence SEQ ID No. 2.
  • a first polynucleotide is considered to be “complementary” to a second polynucleotide when each base of the first nucleotide is paired with the base complementary to the second polynucleotide whose orientation is reversed.
  • the complementary bases are A and T (or A and U), and C and G.
  • an antisense polynucleotide according to the invention has at least 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 300, 400, 500, 1000 or 2000 consecutive nucleotides of the cDNA of ESM-1 of sequence SEQ ID No. 2.
  • a preferred antisense polynucleotide according to the invention consists of a nucleic acid of sequence complementary to the nucleic acid of the cDNA of ESM-1 of sequence SEQ ID No. 2.
  • An antisense polynucleotide constituting an antagonist compound of the ESM-1 protein according to the invention can be prepared by any suitable method well known to those skilled in the art, including by cloning and restriction enzyme action or by direct chemical synthesis. according to techniques such as the phosphodiester method of NARANG et al. (1979) or BROWN et al. (1979), the diethylphosphoramidite method of BEAUCAGE et al. (1980) or the solid support technique described in European patent No. EP-0 707 592.
  • the antisense polynucleotides must have a sufficient length and melting temperature to allow the formation of an intracellular duplex hybrid having sufficient stability to inhibit expression of the ESM-1 mRNA.
  • Strategies for constructing the antisense polynucleotides are notably described by GREEN et al. (1986) and IZANT and WEINTRAUB (1984).
  • antisense polynucleotides are for example those described by SCZAKIEL et al. (1995) or those described in PCT application No. WO 95 / 24.223.
  • Those skilled in the art can advantageously refer to the methods of production and use of antisense polynucleotides inhibiting or blocking the expression of genes associated with the development of cancers, such as the techniques described in US Patent No. 5,582,986 which discloses oligonucleotides antisense for the inhibition of the ras gene, the technique described by HOLT et al.
  • an ESM-1 protein antagonist compound according to the invention can be selected by a person skilled in the art for its ability to inhibit the development of a tumor induced by the ESM-1 protein in vivo.
  • a method for selecting an antagonist compound of the ESM-1 protein comprises the following steps: a) injecting into an animal cells capable of forming tumors in the presence of the ESM-1 protein, said cells being transfected or transformed with a nucleic acid capable of expressing the ESM-1 protein in vivo; b) administering to this animal a candidate antagonist compound of the ESM-1 protein; c) comparing the formation of tumors in a first animal as obtained at the end of step b) and in a second animal as obtained at the end of step a); and d) selecting the candidate compound capable of inhibiting or blocking the formation of tumors in the first animal.
  • the animal used in the above selection process is a non-human mammal, advantageously a rodent, and very preferably a rat, a guinea
  • step e) consisting in sacrificing the first and the second animal.
  • the cell line capable of forming tumors in animals in the presence of the protein ESM-1 is the line
  • HEK 293 ATCC N ° CRL 1573.
  • a protein antagonist compound in another aspect, a protein antagonist compound
  • ESM-1 according to the invention can be selected according to a method making it possible to demonstrate the binding of a candidate compound to the protein ESM-1.
  • a method for selecting a candidate antagonist compound for the ESM-1 protein comprises the following steps: a) providing a polypeptide consisting of the ESM-1 protein or a peptide fragment of this protein; b) bringing said polypeptide into contact with the candidate compound to be tested; c) detecting the complexes formed between said polypeptide and the candidate compound; d) selecting the candidate compounds which bind to the polypeptide consisting of the ESM-1 protein or of a fragment of this protein.
  • fragment of the ESM-1 protein is meant a polypeptide comprising at least 20, preferably at least 30, 35, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140 or 150 consecutive amino acids of the ESM-1 polypeptide of sequence SEQ ID No. 1 and comprising the sequence going from the praline residue in position 133 to the valine residue in position 138 of SEQ ID N ° 1.
  • the invention also relates to a kit or kit for the selection of a candidate antagonist compound of the ESM-1 protein, this kit or kit comprising: a) a purified preparation of a polypeptide consisting of the ESM-1 protein or a fragment of this protein; b) where appropriate, means for detecting a complex formed between the polypeptide and the candidate compound to be tested.
  • the method for detecting a complex formed between the polypeptide derived from the ESM-1 protein and the candidate compound can be carried out by various techniques, such as by microdialysis coupled to an HPLC method as described by WANG et al. (1997) or capillary affinity electrophoresis as described by BOUSH et al. (1997).
  • a candidate compound can be of any kind, and in particular the final product of a combinatorial chemistry process.
  • a candidate antagonist compound of the ESM-1 protein can be selected according to the above method as an expression product of a DNA insert contained in a phage vector according to the technique described by PARMLEY & SMITH (1988) .
  • the DNA inserts encode peptides of 8 to 20 amino acids in length, as described by OLDENBURG KR et al. (1992), VALADON P et al. (1996), LUCAS AH (1994), WESTERINK (1995), FELICI et al. (1991).
  • the recombinant phages expressing a protein capable of binding to the polypeptide consisting of the protein ESM-1 or a fragment thereof is retained and the complex formed between the protein ESM-1 or a fragment of this and the recombinant phage can be subsequently immunoprecipitated by a monoclonal or polyclonal antibody anti-ESM-1.
  • the candidate antagonist compounds of the ESM-1 protein can also be selected in that they bind to the ESM-1 protein, or to a polypeptide fragment thereof, in competition with an antagonist compound of the ESM-1 protein previously selected such as one of the anti-ESM-1 antibodies described above, and in particular the monoclonal antibody secreted by the hybridoma line MEP08 deposited on November 19, 1997 with the CNCM under the access number 1-1941.
  • an antagonist compound of the ESM-1 protein previously selected such as one of the anti-ESM-1 antibodies described above, and in particular the monoclonal antibody secreted by the hybridoma line MEP08 deposited on November 19, 1997 with the CNCM under the access number 1-1941.
  • Such competitive experiences are for example described in the article by BECHARD et al. (2000).
  • Proteins or other molecules of any kind capable of binding to the ESM-1 protein, or a polypeptide fragment of this protein can be selected using affinity columns on which the ESM-1 protein has been immobilized or a fragment of the latter, for example using conventional techniques, including chemical coupling of the protein ESM-1 or a fragment of the latter to the matrix of a column such as agarose, or Affigel®.
  • a solution containing the candidate compound to be tested is brought into contact with the chromatographic support on which the ESM-1 protein or a peptide fragment thereof is immobilized. The compounds retained on the affinity column are selected positively.
  • ESM-1 protein antagonist candidate compound can also be selected using an optical biosensor as described by EDWARDS and LEATHERBARROW (1997).
  • This technique allows the detection of interactions between molecules in real time without requiring the use of labeled molecules.
  • This technique is based on the surface plasmon resonance (SPR for “Surface Plasmon Resonance”). Briefly, the candidate compound to be tested is attached to a surface, such as a matrix of carboxymethyldextran. A light ray is directed on the end of the surface which does not contain the sample to be tested and is reflected by this surface.
  • the SPR phenomenon causes a reduction in the intensity of the reflected light with a specific association between the angle of the reflected light and the wavelength of the light ray.
  • the binding of the candidate compound causes a change in the refractive index of the surface, the change in refractive index being detected as a modification of the SPR signal.
  • Such an optical biosensor detection method can also make it possible to select the candidate compounds which compete with another ligand for binding to the ESM-1 protein or a peptide fragment of the latter.
  • a candidate antagonist compound of the protein ESM-1 includes the compounds capable of inhibiting the binding of an anti-ESM-1 antibody to the protein ESM-1, of inhibiting the binding of factor HGF-SF or else factors FGF-2 and FGF-7 on the protein ESM-1 or a peptide fragment of the latter.
  • the invention relates to a process for the selection of an antagonist compound of the ESM-1 protein, characterized in that it comprises the following steps: a) bringing the ESM-1 protein into contact or a peptide fragment of the latter in the presence of:
  • step a) a candidate compound to be tested; b) in a step separate from step a), but possibly simultaneous with the latter, bringing the ESM-1 protein or a peptide fragment of the latter with an antagonist compound of the ESM-1 protein binding to the ESM-1 protein; c) detecting the respective amount of the antagonist compound of the ESM-1 protein fixed at the end of each of steps a) and b); and d) selecting the candidate compound which competes with the antagonist compound for binding to the ESM-1 protein.
  • an ESM-1 antagonist compound for carrying out the above selection method is an anti-ESM-1 antibody or else a peptide antagonist compound as defined above in the present description.
  • a method for selecting an ESM-1 antagonist compound from a candidate compound comprises the following steps:
  • step 2) administering a compound selected in step 1) to an animal and determining the capacity of this compound to inhibit, in this animal, the development of tumors induced by the ESM-1 protein; 3) select the compounds inhibiting the development of tumors determined in step 2) as antagonist compounds of the protein ESM-1.
  • Step 1) preferably consists of implementing a process for selecting a candidate compound which binds to the ESM-1 protein or a fragment thereof, chosen from the processes detailed in the present description.
  • Step 2) preferably consists of implementing a process for selecting a candidate compound in vivo as detailed in the description. In a particular embodiment of the method, it also comprises a step 4) consisting in sacrificing the animal.
  • Pharmaceutical composition of the invention preferably consists of implementing a process for selecting a candidate compound in vivo as detailed in the description. In a particular embodiment of the method, it also comprises a step 4) consisting in sacrificing the animal.
  • Another subject of the invention consists of a pharmaceutical composition for the treatment and / or prevention of cancer comprising a compound antagonist of the protein ESM-1.
  • composition comprising an antagonist compound of the antibody type or of the peptide type according to the invention.
  • a pharmaceutical composition according to the invention comprises a therapeutically effective amount of an anti-ESM-1 antibody or of a peptide antagonist compound derived from ESM-1, in combination with one or more pharmaceutically compatible vehicles.
  • the pharmaceutical compositions according to the invention include those suitable for topical, oral, rectal, nasal or parenteral administration (including intramuscular, subcutaneous and intravenous) or also in a form suitable for administration by inhalation or insufflation.
  • the pharmaceutical compositions according to the invention can be presented in the form of dosage units and can be prepared by any method well known to those skilled in the art in the pharmaceutical pharmaceutical field. All the methods include a step consisting in associating the agonist compound constituting the active principle of the composition with a liquid vehicle or a finely divided solid vehicle and, if necessary, shaping the product, for example. in the form of tablets or capsules.
  • a pharmaceutical composition according to the invention is preferably presented in the form of dosage units such as capsules, tablets or capsules.
  • the pharmaceutical composition may include a propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gases.
  • a propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gases.
  • the dosing unit can be provided with a valve capable of releasing a given amount of the pharmaceutical composition.
  • the pharmaceutical composition according to the invention may be in the form of a dry powder composition for administration by inhalation or insufflation, for example in the form of a mixture of powder of the antagonist compound and of a suitable base powder, such as lactose or starch.
  • the powder composition can be presented in dosage units, for example in the form of capsules or cartridges from which the powder can be administered using an inhaler or insufflator device.
  • a pharmaceutically compatible solid vehicle of a pharmaceutical composition according to the invention includes substances such as flavoring agents, lubricants, solubilizers, suspending agents, bulking agents, compression aids, binders or agents disintegrants as well as encapsulation materials.
  • the vehicle is a finely divided solid which is mixed with the antagonist compound of ESM-1 also in the finely divided form.
  • the antagonistic active principle of ESM-1 is mixed with a vehicle having suitable compression properties and compacted in the desired shape and size.
  • the powders and tablets preferably contain less than 99% of the active principle.
  • Preferred solid vehicles are for example calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, polyvinylpyrrolidone and ion exchange resins .
  • Liquid vehicles are used to prepare a pharmaceutical composition according to the invention in the form of a solution, a suspension, an emulsion, a syrup, an elixir and a pressurized composition.
  • the antagonist active principle of the protein ESM-1 can be dissolved or suspended in a pharmaceutically acceptable vehicle such as water, an organic solvent, a mixture of the two or pharmaceutically acceptable oils or fats.
  • the liquid vehicle may contain other pharmaceutically acceptable additives such as agents solubilizers, emulsifiers, buffers, preservatives, sweeteners, aromatics, suspending agents, thickening agents, coloring agents, viscosity regulators, stabilizers or osmo-regulators.
  • liquid vehicles for oral and parenteral administration include water, alcohols (including monohydric and polyhydric alcohols such as glycols), oils such as coconut oil or fractionated peanut oil.
  • the vehicle can also be an ester such as ethyl oleate and isopropyl myristate.
  • Liquid pharmaceutical compositions in the form of sterile solutions or suspensions can be used for intramuscular, intraperitoneal or subcutaneous injection.
  • a pharmaceutical composition according to the invention comprises from 1 to 1000 mg of antagonist compounds of the ESM-1 protein per dosage unit, and preferably from 10 to 500 mg of the antagonist compound of the ESM-1 protein per unit of dosage dosage.
  • the present invention also relates to a method of treatment and / or prevention of cancer comprising a step during which a pharmaceutical composition as defined above is administered to a patient in need of such treatment.
  • composition comprising an antagonist compound of the ESM-1 protein of the antisense polynucleotide type.
  • compositions containing a therapeutically effective amount of a protein antagonist compound ESM-1 of the antisense polynucleotide type as defined in the present description as well as methods of treatment and / or prevention of a cancer comprising the administration to a patient in need of such treatment of a pharmaceutical composition comprising an antisense polynucleotide as defined above.
  • An antisense oligonucleotide according to the invention can be administered by any means, whether local or systemic.
  • the local administration of an antisense polynucleotide of the invention can be carried out by the administration of the antisense polynucleotide directly at the level of the tumor or at the level of the tissue surrounding the tumor and allowing the oligonucleotide to migrate and, where appropriate, to enter the tumor cells.
  • antisense polynucleotides can be injected using a syringe. The injection can be intramuscular, intravenous, intraperitoneal or subcutaneous.
  • the antisense polynucleotide can be administered to the liver through the hepatic portal vein system. Likewise, the antisense polynucleotide can be administered to the lung using an inhalation device.
  • an antisense polynucleotide can be administered systemically after their insertion into an expression vector.
  • expression vector encompasses a plasmid, a virus or another vehicle known in the art for ensuring the expression of an antisense polynucleotide.
  • vectors suitable for the recombinant expression of an antisense polynucleotide may advantageously use pMSXND vectors described by LEE and NATHANS (1988), eukaryotic viral vectors, such as those described by GLUZMAN (1982), or adenoviruses and adeno-associated viruses such as those described in US patents 5, 173,414 and 5,354,678 or also an expression system including the expression vector described by MOXHAM et al. (1993).
  • the expression vector contains a promoter allowing the production of the antisense polynucleotide in an animal, preferably a mammal, and more preferably in humans, such as the polyhedrin promoter.
  • the expression vector can be adapted to the targeted expression of the antisense polynucleotide at the tumor site, for example by placing the nucleic acid coding for the antisense polynucleotides under the control of a promoter specific for certain cells, such as epithelial cells or endothelial cells.
  • a promoter is the designated viral promoter NuNTV which is useful specifically in the treatment of breast cancer.
  • the therapeutically effective amount of an antisense polynucleotide of the invention can be determined as the amount necessary for a significant reduction in the translation of the protein ESM-1 at the systemic or local level.
  • the concentration of the therapeutically effective antisense polynucleotide varies with the choice of the form of administration.
  • the dosing unit consisting of a syringe containing an effective amount of the antisense polynucleotide.
  • An effective amount of the antisense polynucleotide for systemic administration is between 0.01 mg / kg to 50 mg / kg administered once or twice a day.
  • a therapeutically effective amount of an antisense polynucleotide according to the invention included in a pharmaceutical composition is generally between 10 4 and 10 11 molecules of antisense polynucleotide per administration and preferably between 10 5 and 10 10 DNA molecules per administration.
  • assay protocols can be used depending on (i) the individual ability of the antisense polynucleotide to inhibit expression of the ESM-1 protein, (ii) the severity or extent of the disease, or (iii) behavior pharmacokinetics of the antisense polynucleotide used.
  • the antisense polynucleotide can be associated with a pharmaceutically compatible vehicle or with an excipient.
  • excipients include bulking agents, binders, disintegrants, lubricants, depending on the nature of the administration and the dosage forms.
  • Preferred dosage forms consist of liquid solutions, preferably physiologically compatible buffers such as HANK's or RINGER solutions.
  • the antisense polynucleotides according to the invention can be formulated in solid form and then redissolved or resuspended immediately before use. According to encompasses lyophilized forms and liposomes containing such antisense polynucleotides.
  • An antisense polynucleotide of the invention can also be administered systemically by the transmucosal, transdermal or oral routes.
  • penetrating agents can be used in the formulation such as bile salts or fusidic acid derivatives.
  • the present invention also relates to a method of treatment and / or prevention of cancer comprising a step of administration to a patient, in need of such treatment, a pharmaceutical composition as defined above comprising a compound ESM-1 antagonist of the antisense polynucleotide type .. ...
  • any of the pharmaceutical compositions of the invention as defined above and comprising a therapeutically effective amount of an ESM-1 protein antagonist compound is useful in the prevention and / or treatment of cancer.
  • a pharmaceutical composition according to the invention is useful for the prevention and / or treatment of cancers such as cancers of the respiratory tract, bronchopulmonary cancers, breast cancers, colon cancers and renal cancers as well as cancers of the digestive tract.
  • cancers such as cancers of the respiratory tract, bronchopulmonary cancers, breast cancers, colon cancers and renal cancers as well as cancers of the digestive tract.
  • FIG. 1 illustrates immunoblotting gels (“Western-Blot”) and stains of ESM-1 on SDS-PAGE gel. Each immunoblock gel was revealed with the anti-ESM-1 MEP14 monoclonal antibody. The second anti-mouse antibody labeled with horseradish peroxidase has been affinity purified and gives negative results when used alone.
  • Figure 1A ESM-1 protein immunoblot gel of different cell types expressing this protein.
  • ESM-1 Immunoprecipitation of ESM-1 from culture supernatants of cells SVI (1), 293-ESM (2) and CHO-ESM (3) was carried out with the antibody MEP19 when indicated, or with a control antibody.
  • the arrows indicate the specific band of ESM-1.
  • the native form of ESM-1 is represented by a diffuse band around 50 kD.
  • FIG. 1B Absence of detection of the purified protein ESM-1 with coomassie blue.
  • FIG. 1C Detection of the protein ESM-1 purified with alkian blue.
  • FIG. 2 illustrates the apparent molecular weight of the peptide and glycan bodies of ESM-1.
  • FIG. 1A Analysis by mutation of the O-glycosylation binding site.
  • the wild ESM-1 protein (WT for “Wild-Type”), the ESM-1 mutants T120A and S137A) as well as the negative controls (MOCK) were transfected into 293 cells and the cell culture supernatants as well as the cell lysates were analyzed by immunoblotting (Western-Blot) using the monoclonal antibody MEP14. The arrows indicate the specific bands.
  • FIG. 2B Effect of proteinase K treatment on ESM-
  • the ESM-1 protein purified from SVI (1) and 293-ESM (2) cells was digested with proteinase K and loaded onto a 15% SDS-PAGE gel.
  • the upper arrow indicates the wild form of the untreated ESM-1 protein and the lower arrow indicates the proteinase K digested ESM-1 protein.
  • Figure 3 illustrates the effects of specific chondroitinases on ESM-1.
  • FIG. 3A Treatment of purified wild protein ESM-1 with chondroitinase ABC.
  • the secreted ESM-1 protein was purified by ion exchange chromatography, followed by immunoaffinity chromatography from supernatants from SVI (1), 293-ESM (2) cell cultures and from of human plasma (3), then digested or not by chondroitinase ABC. 50 ng of the digested protein were loaded onto a 15% SDS-PAGE gel and then analyzed by immunoblotting (Western-Blot). The upper arrow indicates undigested forms of ESM-1 and the lower arrow indicates digested forms of ESM-1.
  • Figure 3B Treatment of purified wild-type ESM-1 protein with chondroitinase B.
  • ESM-1 protein purified from culture supernatants of SVI (1) and 293-ESM (2) cells were digested or not by chondroitinase. Proteins were loaded onto a 15% SDS-PAGE gel.
  • the upper arrow indicates the different forms of ESM-1 no digested around 50 kD and the lower arrow indicates the different forms of the digested ESM-1 protein, around 22 kD.
  • Figure 3C Treatment of purified wild protein ESM-1 with chondroitinase AC.
  • the protein ESM-1 purified from culture supernatants of cells HUVEC (1) and 293-ESM (2) have been fired ??? with chondroitinase AC and were loaded onto a 15% SDS-PAGE gel.
  • the upper arrow indicates the different forms of undigested ESM-1 around 502 kD and the lower arrow indicates the different digested forms of the ESM-1 protein around 22 kD.
  • Figure 3D Treatment of wild ESM-1 protein. purified with chondroitinase C.
  • the protein ESM-1 purified from culture supernatants from HUVEC (1) and 293-ESM (2) cells were digested or not by chondroitinase C and were loaded onto a 15% SDS-PAGE gel.
  • the upper arrow indicates the different undigested forms of ESM-14 around 50 kD and the lower arrow indicates the different digested forms of the ESM-1 protein around 22 kD.
  • FIG. 4 illustrates the effects of the purified wild-type ESM-1 protein on the coagulation time in the presence of thrombin.
  • the delay and reduction in thrombin formation can be noted for the heparin plasma as well as for the other four curves (plasma rich in platelet or PRP: empty diamond; plasma rich in platelet + ESM-1 at 0.2 mg / ml : full square; plasma rich in platelet + ESM-1 0 0.5 mg / ml: solid triangle; plasma rich in platelet + ESM-1 at 1 mg / ml: full circle; plasma rich in platelet + heparin: empty circle) .
  • FIG. 5 illustrates the biological activity of the proteoglycan ESM-1 on the proliferation of 293 cells induced by the HGF / SF factor.
  • the stimulation of the incorporation of 3 H-thymidine by the 293 cells induced by the HGF / SF factor was studied.
  • the cells were seeded at 1 ⁇ 10 4 cells per well in DMEM medium added transferin and insulin and HGF / SF at 50 ng per ml before adding different molecules.
  • the bars represent the percentage increase in the incorporation of 3 H-thymidine (mean +/- SEM of samples in triplicate from a representative experiment) in the presence of the indicated additions of serum, of the different forms of ESM-1 to 2.5 mg / ml and decorin at 2.5 mg / ml.
  • the background noise of incorporation of 3 H-thymidine in the presence of HGF / SF was generally between 7,000 and 8,000 cpm per well. The results presented are similar to those obtained in three other separate experiments.
  • FIG. 6 illustrates a study of twelve responses of the different forms of ESM-1 and decorin on the mitogenic activity induced by the HGF / SF factor.
  • ESM-1 / WT empty square
  • ESM / S137A solid circle
  • GAG chain derived from the protein ESM / WT solid square
  • decorin empty circle
  • FIG. 7 illustrates the tumorigenicity of the protein ESM-1.
  • Two batches of more than 10 mice received HEK control cells or HEK cells transfected with a vector coding for the cDNA of the wild-type ESM-1 protein (ESM / WT).
  • ESM / WT wild-type ESM-1 protein
  • FIG. 7A the percentage of macroscopically visible tumors at the eighth week at the injection site and whose tumor volume is greater than 1 cm 3 is represented on the ordinate.
  • FIG. 7B illustrates the kinetics of appearance of tumors in mice having received transfected HEK cells expressing the glycosylated protein ESM-1 (ESM / WT). The number of weeks after the injection of the cells is shown on the abscissa. The ordinate shows the average tumor volume, expressed in cm 3 .
  • FIG. 8A represents the serum level of protein ESM-1 found in the two batches of mice, at the eighth week following the injection of the cells. On the abscissa are shown respectively the batch of mice having received the control HEK cells and the batch of mice having received the HEK cells expressing the glycosylated protein ESM-1 (ESM / WT). The serum ESM-1 level found, expressed in nanograms / ml, is represented on the ordinate.
  • FIG. 8B illustrates the kinetics of the serum levels of ESM-1 measured by ELISA, for the mice of the batch having received HEK cells transfected with a DNA coding for the glycosylated protein ESM-1 (ESM / WT).
  • the abscissa shows the number of weeks following the injection of the transfected cells.
  • the serum ESM-1 protein level, expressed in nanograms / ml, is represented on the ordinate.
  • FIG. 9 illustrates the tumorigenic activity of different forms of the protein ESM-1.
  • FIG. 9A illustrates the appearance of tumors in different batches of mice, the mice having respectively received the HEK control cells, the HEK cells transfected with a cDNA coding for the glycosylated protein ESM-1 (ESM / WT), the cells transfected with the non-glycosylated ESM-1 protein (ESM / S137A) and the HEK cells transfected with a cDNA coding for the ESM-1 protein mutated at positions 134 and 135 (ESM / F1 15A, F116A).
  • the ordinate shows the percentage of macroscopically visible tumors at the eighth week at the injection site, the tumor volume of which is greater than 1 cm 3 .
  • FIG. 9B illustrates the serum level of ESM-1 in the different identical batches of mice.
  • the serum ESM-1 level, expressed in nanograms / ml, is represented on the ordinate.
  • FIG. 10 illustrates the inhibitory effect of the monoclonal antibody MEP08 on the pro-tumor activity of the protein ESM-1.
  • the injection of MEP-08 antibodies increases the survival of the mice of the HEK ESM / WT group.
  • the MEP-08 monoclonal antibodies are injected intraperitoneally at a dose of 400 ⁇ g from the second following inoculation of the HEK / ESM-WT cells. The injections are repeated weekly for 12 weeks.
  • a control antibody, MEP-14 is used under the same conditions.
  • the mice are sacrificed when their tumor volume is greater than 6 cm 3 , (n> 8 mice in each group). The figure represents the percentage of live mice in each of the groups.
  • CHO cells were cultured in a MAM ⁇ culture medium (Gibco BRL, Life Technologies, France) supplemented with 10% fetal calf serum.
  • Human endothelial cells transfected with the SV40 virus, the SV1 cells described by LASSALLE P et al. (1992) were cultured in RPMI 1640 medium containing 2 mM L-glutamine and 10% fetal calf serum.
  • Human embryonic kidney cells, cells of line 293, were cultured in DMEM medium from Dulbecco with 10% fetal calf serum.
  • the human embryonic renal cells, cells of line 293, used for the proliferation test were cultured in medium of EAGLE modified by Dulbecco (Gibco BRL) supplemented with insulin at 10 mg / ml and with transferin at 10 mg / ml.
  • Proteinase and chondroitinase ABC are marketed by Boehringer Mannheim.
  • Chondroitinases B, AC and C are marketed by Sigma Human HGF / SF factor is marketed by R & D and decorin is marketed by Sigma.
  • Anti-ESM-1 monoclonal antibodies were produced and purified as described by BECHARD et al. (2000).
  • the complete cDNA encoding ESM-1 was directed, purified and inserted into the expression vector pcDNA3 (marketed by Invitrogen) between the Xhol and Hindllll sites.
  • the vector constructs were transfected into CHO and 293 cell lines in the presence of lipofectamine (GIBCO BRL), then selected on G418 (1000 ⁇ g / ml for the CHO line and 300 ⁇ g / ml for the line 293).
  • Stably transfected cell lines were obtained by limiting dilution and the cells thus selected were designated CHO-ESM and 2936-ESM respectively
  • the serine residue at position 137 (SEQ ID No. 1) and the threonine at position 120 (SEQ ID No. 1) were substituted with an alanine residue.
  • O-glycosylation mutants were produced by PCR using the Quick kit
  • the mutated cDNAs were confirmed by sequencing (ABI prism 377 sequencer from the company Applied Biosystems. Then the 293 cells were transfected with the vectors into which the mutant cDNAs were inserted to obtain transient transfectants and stable transfectants, respectively the lines. 293-ESM / S 137A and 293-ESM / T120A.
  • the cell culture supernatants were adjusted to pH8, then passed over a DEAE-Sepharose column (Pharmacia), washed with a 50 mM Tris buffer (pH8), 0.2 MNaCI, then eluted in a 50 mM Tris buffer (pH8 ), 0.8 M Nacl.
  • the eluates were adjusted to 50 mM Tris (pH8), 0.5 MNaCI and passed through an affinity column.
  • the affinity column consists of anti-ESM-1 monoclonal antibodies (produced by the hybridoma line MEC4) immobilized on an Affigel Hz hydrazide gel, according to the manufacturer's recommendations (Biorad).
  • the ESM-1 protein was eluted with a 3M solution of MgCI 2 , concentrated and dialyzed against the same buffer on an ultrafree device 30 ( millipore).
  • the eluted material was then quantified by immunodetection with anti-ESM-1 antibodies, and checked on SDS-PAGE using a staining with coomassie blue or alkian blue.
  • the purification of the ESM-1 protein from human plasma was carried out according to the following protocol.
  • ESM-1 The non-glycosylated form of ESM-1 (ESM / S137A) was purified in a single step by chromatography and immunoaffinity. The degree of purity of the glycosylated protein ESM-1 (ESM / WT) and of the serine 137 non-glycosylated protein (ESM / S137A) was checked by FPLC. The purified material is free of endotoxins, as evidenced by the results of the limulus amebocyte lysate test (BlOwhitaker).
  • ESM-1 The size of the different forms of ESM-1 was determined by immunoprecipitation and immuno-imprinting from cell culture supernatants and cell lysates.
  • the cells were lysed in a buffer containing 0.5% of NP40, a cocktail of anti-proteases (Boehringer Mannheim, Germany) in PBS for 30 minutes at 4 ° C with shaking. Then, the lysates were centrifuged at 10,000g for 15 min to obtain clarified cell lysates.
  • the culture supernatants were filtered through a filter having a pore diameter of 0.45 mm.
  • 1 ⁇ g of the ESM-1 monoclonal antibody produced by the MEP19 hybridoma line or 1 ⁇ g of anti-ICAM-1 monoclonal antibody (clone 164B) was added to the clarified lysate or to the cell culture supernatant and incubated for one overnight at 4 ° C with stirring.
  • the beads were resuspended in 20 and 40 ⁇ l of SDS-PAGE buffer for 5 min, centrifuged, and the supernatants were analyzed.
  • the samples were subjected to SDS-PAGE gel electrophoresis, then transferred to a nitrocellulose membrane according to standard procedures.
  • the membranes were incubated for one hour with a monoclonal antibody ESM-1 produced by the hybridoma line MEP14 at 1 ⁇ l, washed and then incubated for I hour with a secondary anti-mouse Fc antibody conjugated to horseradish peroxidase ("Horse Radish peroxidase”) (sold by the company Sigma).
  • Horse Radish peroxidase horseradish peroxidase
  • washes were carried out before revelation using the ECL detection kit sold by the company Amersham.
  • the purified ESM-1 protein was subjected to electrophoresis on an SDS-PAGE gel, then electrotransfered onto a polyvinylidene difluoride (PVDF) membrane sold by the company Millipore, then stained. using 0.1% coomassie blue.
  • PVDF polyvinylidene difluoride
  • the protein band at 50 kD was excised from the membrane and the N-terminal sequence was carried out by degradation of automated EDMAN on a protein sequencer of the ABI 473A type.
  • ESM-1 ratio equal to 1: 50 (weight: weight) in a 10 mM Tris buffer, pH8, in the presence or in the absence of 0.1% SDS at 56 ° C for 3 hours.
  • BSA bovine serum albumin
  • the purified ESM-1 protein was digested with several chondroitinases: ABC chondroitinases (0.5 unit / mg in 100 mM TrisHCI buffer, pH 8, 30 mM acetate sodium, pH 5.2 at 37 ° C for 45 min), chondroitinase B (200 units / mg in 20 mM Tris-HCl buffer, 50 mM Nacl, 4 mM CaCl 2 , 0.01% BSA, pH 7.5 at 25 ° C for two hours), chondroitinase AC (one unit per ml in 250 mM Tris Hcl buffer, 75 mM sodium acetate, pH 7.3 at 37 ° C for two hours) chondroitinases C (80-120 units / ml in 50 mM Tris HCl buffer, pH 8 at 25 ° C for 3 hours). The samples were analyzed by immunoblotting.
  • ABC chondroitinases 0.5 unit / mg in 100 mM TrisHCI buffer, pH
  • the control plasma poor in platelets was prepared from blood in the presence of the anticoagulant sodium citrate (30 mM), by centrifugation at 2500 g for 15 min. All reagents are marketed by STAGO Diagnostica (France). Three parameters were evaluated, by adding the ESM-1 protein, buffer or heparin to the plasma poor in platelets: a) APTT (for “Activated Partialo Thromboplastin Time”): this parameter explores the intrinsic pathway of blood coagulation (FI, Wire, FV, FVIII, FIX, FX, FXI, FXII). The deficit or inhibition of one of these factors increases the coagulation time of the PPP reactive mixture, cephalin, activator, CACI 2 .
  • TCT for “Thrombin Clotting Time”: this parameter is analyzed on a mixture of plasma poor in platelets (PPP) in the presence of thrombin. With a standard concentration of thrombin, the plasma coagulation time is constant. Defects in fibrin formation induce an increase in clotting time.
  • anti-Xa activity the anti-Xa activity of heparin or of other inhibitors acting on the factor FXa is detected by a competitive test.
  • the studied sample PPP + ESM-1, + buffer or + heparin
  • factor Fxa a chromogenic substrate specific for factor Fxa. The final coloration is inversely proportional to the concentration of inhibitor.
  • This sensitive global test can detect plasma or platelet defects inducing a delay or a reduction in the generation of thrombin.
  • a platelet rich plasma PRP
  • PRP platelet rich plasma
  • the thrombin generation test was carried out, for each of the subjects, in samples in the absence of ESM-1, with unfractionated calcium heparinate (0.5 IU of anti-Xa / ml) or with 0.2mg / ml, 0.5 mg / ml and 1 mg / ml ESM-1 (final concentration).
  • the ESM-1 protein was added 10 min before the test. At 37 ° C, 1 ml of plasma was mixed with 1 ml of CaCl 2 and the stopwatch was started. 0.1 ml aliquots were taken up from the reaction mixture every minute for 1 min.
  • the clot formed in the reaction mixture is regularly removed.
  • the aliquots were mixed with 0.2 ml of fibrinogen (Sigma, 4/1000 in Owren buffer at 37 ° C and the coagulation time was measured for each of the aliquots).
  • the thrombin formed in the reaction mixture acts on the forminogens, inducing the formation of fibrin.
  • the coagulation activity was maximum between 4 and 8 min and then decreased due to the neutralization of the thrombin by the anti-thrombin.
  • ESM-1 glycosylated (ESM / WT) and non-glycosylated ESM-1 (ESM / S137A) purified in 50 mM Tris buffer, pH 8.5, 0.5 MNAcl were separated by liquid chromatography on a Superdex 200 column (for ESM / WT) or Superdex 75 column (for ESM / S137A) sold by Pharmacia, using the Biorad Biologie Chromatography System with a flow rate of 1 ml / min.
  • ribonuclease A from bovine pancreas, 13.7 kD
  • ovalbumin 43 kD
  • albumin bovine serum, 67 kd
  • aldolase rabbit muscle, 158 kD
  • ferritin horse spleen, 440 kD
  • thiroglobulin bovine thyroid, 669 kD
  • the molecular weight standards were separated using a buffer identical to that used for the ESM-1 proteins and the separation was carried out immediately after the separation of the ESM / WT and ESM / S137A proteins.
  • ESM-1 protein was detected using a specific enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • ESM-1 Post-translational modifications of the secreted form of the ESM-1 protein produced by endothelial cells and by established cell lines.
  • the protein ESM-1 was purified from the cell line 293-ESM.
  • the N-terminal sequence of the 50 kD form indicated that the signal peptide of 19 amino acids was cleaved at the predicted site, resulting in a mature ESM-1 polypeptide of 165 amino acids starting at the tryptophan residue in position 20 of the sequence SEQ ID No. 1, the N-terminal sequence being "WSNNYAVD-P".
  • ESM-1 was immunoprecipitated from the cell culture supernatants of HUVEC, SV1, 293-ESM and CHO-ESM cells, then analyzed by immunoblotting.
  • ESM-1 The molecular weight found is more important than the predicted molecular weight. This result suggested that the secreted form of ESM-1 had undergone post-translational modifications. The fact that the purified ESM-1 protein was better stained on the SDS-PAGE gel with alkian blue than with coomassie blue suggested that ESM-1 was glycosylated (FIGS. 1B, 1C) rather than oligomerized across the bridge disulfide, since reducing conditions do not change the apparent molecular weight of ESM-1.
  • the serine residue at position 137 (SEQ ID No. 1) is the O-glycosylation site of ESM-1.
  • a computer analysis of the potential glycosylation sites made it possible to identify three putative sites of O-glycosylation, respectively for the serine in position 16, on the threonine in position
  • the threonine residue at position 120 and the serine residue at position 137 have been mutated and replaced by an alanine residue. These mutants were transiently expressed in 293 cells.
  • the ESM-1 protein was then immunoprecipitated from cell lysates and culture supernatants, and analyzed by immunoblotting.
  • ESM / T120A protein migrates at 50 kD, at a position similar to the apparent molecular weight of the wild form of ESM-1 (ESM / WT), as shown in Figure 2A.
  • ESM / S137A protein migrates to 22 kD corresponding to the intracellular form of ESM-1 ( Figure 2A), a molecular weight compatible with the predicted molecular weight of ESM-1.
  • glycosaminoglycan (GAG) of ESM-1 In order to determine the length of the glycosaminoglycan (GAG) of ESM-1, the peptide body of ESM-1 was completely digested by proteinase K.
  • Proteinase K treatment induces a change in molecular weight from 50 kD to 25-30 kD (Figure 2B). These results show that the band of apparent molecular weight at 50. kD is compatible with the presence of a 22 kD polypeptide which is glycoconjugated on the serine at position 137 by a GAG chain with an average size of 25-30 kD.
  • the profile is similar with the protein ESM-1 purified from 293-ESM cells as well as from the human endothelial cell line SVI. Because the protein ESM-1 circulates in the blood, not a also studied the behavior of the protein ESM-1 purified from human plasma. The results make it possible to observe a single main band of 50 kD, which has a molecular weight of 22 kD after treatment with chondroitinase ABC, as for all the other cell lines studied (FIG. 3A). Thus, the ESM-1 protein is a soluble proteoglycan containing a single chain of chondroitin sulfate.
  • the GAG chain of ESM-1 is a heterogeneous chain of chondroitin / dermatan sulfate.
  • chondroitinases B Treatment with chondroitinase B reduces the apparent molecular weight from 50 kD to 22 kD ( Figure 3B).
  • GAG chain of ESM-1 contains different composite units containing a type of amino sugar, N-acetylgalactosamine, coupled to differently sulphated iduronic or glucuronic acid.
  • ESM-1 protein is secreted as a proteoglycan of the chondroitin / dermatan sulfate type by endothelial cells and because dermatan sulfate has effects on the generation of thrombin in vitro DELORME et al., (1996) and on coagulation, the potential anticoagulant activity of ESM-1 has been verified on the parameters APTT, TCT, anti-Xa activity and on the generation of thrombin.
  • the APTT, TCT and anti-Xz activity parameters are similar for platelet-poor plasma (PPP) with the buffer or with the protein ESM-1. In the positive controls, the APTT, TCT and anti-Xa activities are greater for the PPP in the presence of heparin.
  • the ESM-1 protein has no inhibitory effect on the thrombin generation test: there is no difference according to the concentrations of 0.2 mg / ml, 0.5 mg / ml and 1 mg / ml of ESM-1 relative to the buffer control, while heparin induces a delay in the formation of thrombin (FIG. 4).
  • the proliferation-stimulating activity was determined by measuring the incorporation of 3 H thymidine by the 293 cells.
  • the 293 cells were seeded at the concentration of 1 ⁇ 10 4 cells per well in 96-well microplates of the TPP type and were maintained for 24 hours in the DMEM culture medium supplemented with transferin and insulin.
  • the recombinant human HGF / SF was diluted in PBS containing 0.1% bovine serum albumin and added with water from 3 identical wells in order to obtain a final concentration of 50 ng / ml.
  • the recombinant proteins ESM / WT, ESM / S137A, the purified GAG chain derived from ESM-1 and decorin were added alone or in combination with the factor HGF / SF at doses ranging from 1 ng / ml to 2.5 ⁇ g / ml, simultaneously with the addition of HGF / SF.
  • HGF / SF alone at 50 ng / ml induces proliferation of 293 cells at a level equal to approximately 45% of the proliferation induced by serum, while the protein ESM / WT alone did not stimulate the proliferation of 293 cells.
  • the effect of the protein ESM / WT was compared with the effect of decorin, another proteoglycan of the chondroitin sulfate / dermatan sulfate type, on the mitogenic activity of the HGF / SF factor.
  • decorin does not show any activity for increasing the proliferation of 293 cells induced by the HGF / SF factor (FIGS. 5, 6).
  • ESM-1 The non-glycosylated form of ESM-1 is incapable of inducing proliferation of 293 cells, whether in the presence or in the absence of the factor HGFSF (FIG. 5), even when it is used in high concentration.
  • the GAG chain purified from ESM-1 considerably increases the proliferation of 293 cells induced by the HGF / SF factor, with an increase factor close to 96.6%, compared to the HGF / SF factor alone (FIG. 5).
  • the pro-mitogenic effect of the GAG chain is less than that observed with the wild form of the protein ESM-1, but this effect is nevertheless dependent on the dose of GAG chain added (FIG. 6).
  • the HGF / SF factor is expressed during the critical early periods of human organogenesis of 6 to 13 weeks of gestation. Organs that express the HGF / SF gene are notably the liver, the metanephric kidney, the intestine and the lung, each of these organs developing by inductive interaction between the mesenchyme and the epithelium.
  • the HGF / SF factor is an important factor in human renal multicystic dysplasia (TAKAYAMA et al., 1997) as well as in the appearance of malformation and hyperproliferation in the tubules.
  • ESM-1 protein significantly increases the proliferation of embryonic kidney cells in the presence of HGF / SF while the non-glycosylated form and of the ESM-1 protein has no effect.
  • GAG chain isolated from the ESM-1 protein is capable of mimicking the effects of the glycosylated protein ESM / WT.
  • the protein ESM-1 is selectively detected in the distal tubules, the result which can be associated with the observation of the preferential localization of the HGF / SF factor in the same part, of the nephron in situations of renal multicystic dysplasia human (WEIDNER et al., 1993).
  • WEIDNER et al. 1993
  • These results indicate an application of the protein ESM-1 in pathological disorders dependent on the factor HGF / SF, which has also been shown to be associated in the development of cancers of the breast (RAHIMI et al., 1998), of the kidney (NATALI et al, (1996)) and lung (OTSUKA et al., 1998) but also in malignant melanomas (SIEGFRIED et al., 1998).
  • the HGF / SF factor is likely to promote the extension of hyperplasia and to generate cells with an invasive phenotype.
  • the ESM-1 protein is likely to be involved in these phenomena of deregulated mitogenic activities of the HGF / SF factor.
  • the native form of the ESM-1 protein produced by the CHO cell line transfected with a vector d the expression containing a DNA insert coding for the protein ESM-1 has been purified.
  • the ESM-1 cDNA is inserted into the eukaryotic expression vector pcDNA3 (In vitrogen) and then transfected into CHO cells with lipofectamine (GIBCO) according to the manufacturer's recommendations. 48 hours after transfection, the cells are subcultured in the presence of a selection agent (G418, Gibco) at a dose of 1000 micrograms / ml). After two weeks of selection, the G418-resistant CHO cells are given by limiting dilution. Clones expressing ESM-1 are then selected and called CHO-ESM (deposited at the CNCM). For production, CHO-ESM cells are cultured in suspension in a conditioned medium without fetal calf serum (CHO SFM II medium, Gibco).
  • the supernatant is adjusted to pH 8 and weighed on a DEAE-sepharose column (Pharmacia).
  • the column is washed with a 50 mM Tris buffer, pH 8, 0.2 M NaCl.
  • the ESM-1 molecule is eluted in a 50 mM Tris buffer, pH 8, 1 M NaCl.
  • the eluate is then diluted 1: 4 in a 50 mM Tris buffer, pH 8 and incubated in the presence of anti-ESM-1 monoclonal antibody (MEC4) immobilized on agarose (Biorad). After an overnight incubation at 4 ° C. with agitation, the agarose beads are washed with the 50 mM Tris buffer, pH 8, 0.2 M NaCl.
  • ESM-1 is elected with 3M MgCI 2 .
  • the eluate is concentrated and dialyzed in the 50 mM Tris buffer, pH 8, 0.5 M NaCl and stored at -70 ° C.
  • Balb / C mice were immunized by injection of 10 ⁇ m of recombinant protein ESM-1 purified by mouse, according to a standard immunization protocol in the presence of Freund's adjuvant.
  • Hybridoma cells secreting anti-ESM-1 monoclonal antibodies were obtained by fusion, screening and subcloning according to the technique described by BECHARD et al. (2000).
  • hybridoma Five cell clones of hybridoma were obtained and were generically designated MEC ("Mouse Monoclonal Antibody to ESM-1 produced by CHO Cells"). Four of the hybridomas selected are isotypes lgG1, k respectively the hybridomas designated MEC4, MEC5, MEC15 and MEC36.
  • hybridomas is the IgM.k isotype, the MEC11 hybridoma.
  • the cell clones of hybridomas were cultured in culture medium in the absence of serum and the anti-ESM-1 antibodies were purified by column chromatography on protein G-Sepharose sold by the company Pharmacia (UPSALA, Sweden) .
  • Directed mutagenesis was carried out with the kit marketed by the company STRATAGENE under the reference “Site-directed quick mutagenesis kit", which was used according to the manufacturer's recommendations.
  • a pair of round-trip primers of strictly complementary sequences are synthesized, these primers comprising the nucleotides coding for the mutated amino acid (s), or the complementary nucleotides, these nucleotides being located at the center of the sequence of the primers which also include approximately 10 to 15 consecutive nucleotides complementary to the sequence to be amplified both 5 'and 3' of the central nucleotides.
  • the amplified polynucleotides encoding the mutated ESM-1 protein are inserted into the vector pCDNA3.
  • Primer go: 5'-GCC TGA AAT TCC CCG CCT TCC AAT ATT CAG-3 '(SEQ ID N ° 3).
  • the cell line named HEK ESM / WT was transfected stably with the cDNA encoding the wild form of ESM-1 (ESM / WT).
  • ESM / WT The cell line named HEK ESM / WT was transfected stably with the cDNA encoding the wild form of ESM-1 (ESM / WT).
  • ESM / WT Four other cell lines are obtained by transfection with cDNA encoding purified forms of ESM-1 obtained by site-directed mutagenesis from the wild form.
  • the first of these, called HEK ESM / S137A expresses the mutated, non-glycosylated protein ESM-1, where an alanine is substituted for serine 137, the major site of O-glycosylation.
  • the other three lines expressing a glycosylated form of ESM-1 whose protein part is mutated.
  • ESM-1 HEK ESM / WT
  • ESM-1 HEK ESM / S137A
  • mice used are of the SCID (Severe Combined IMMUNO Deficiency) type. More specifically, they are C.B.17 Scid / scid mice supplied by the animal facility of the Institut Pasteur in Lille. These mice have an autosomal recessive mutation in their recombination system (Blunt., 1995). This mutation results in the production of immunoglobulins and non-functional T cell receptors (TcR) and B 5BcR). As a result, they do not have functional T and B lymphocytes; these mice therefore tolerate non-self and represent a model. of choice for the development of xenogenic tumors.
  • the SCID mice used are male and young mice since they are 3 to 5 weeks old.
  • anti-ascialo GM-1 antibodies 100 ⁇ g per mouse diluted in 200 ⁇ l of RPMI
  • rabbit polyclonal antibodies (Wako Pure Chemical Industries, Ltd) directed specifically against the asialo GM-1 antigen expressed by NK cells.
  • mice Four batches of mice (10 to 15 mice per group) anesthetized with ether are then injected, subcutaneously at the back. Each mouse receives 1 million cells diluted in 200 ⁇ l of RPMI. The injection of these cells defines the first day of the experiment (Jo). For each mouse, macroscopic monitoring of the injection site in search of the appearance of a possible tumor as well as a measurement of the body weight are carried out weekly. From the 5th week, the mice are bled (approximately 500 ⁇ l per mouse), once a week, in order to measure the serum levels of ESM-1 by a ELISA test (BECHARD D et al., 2000). For each mouse, an anatomo-pathological examination is carried out.
  • HEK cells were transfected with a vector having an insert containing the cDNA coding for the glycosylated wild-type protein ESM-1, designated ESM / WT.
  • HEK cells are injected subcutaneously into 5 week old SCID mice. Each mouse receives. previously injected anti-asialo GN-1 antibody intraperitoneally. The percentage of tumors with a volume greater than 1 cm 3 observed in the mice at the eighth week following the injection of the transfected HEK cells was analyzed.
  • FIG. 7A it is observed that the injection of HEK control cells does not induce the appearance of tumors in the mouse.
  • HEK cells transfected with DNA encoding the glycosylated protein ESM-1 induce many macroscopically visible tumors, of which approximately 95% of them have a tumor volume greater than 1 cm 3 .
  • FIG. 7B illustrates the kinetics of appearance of tumors in mice which have received HEK cells transfected with DNA coding for the glycosylated protein ESM-1. It can be observed that the average tumor volume, expressed in cm 3 , increases continuously from the fourth week following the injection of the transfected HEK cells.
  • the experimental results presented in FIG. 7 clearly demonstrate that the glycosylated protein ESM-1 has a pro-tumor activity.
  • ESM-1 protein serum levels were also measured in mice having received control HEK cells and in mice having received HEK cells transfected with the cDNA encoding protein ESM-1. The results are shown in Figure 8.
  • FIG. 8A show that the ESM-1 protein is not found in the sera of the mice having received the HEK control cells. On the contrary, a serum level of 40 to 50 nanograms per ml is found in mice having received HEK cells transfected with the cDNA coding for the protein ESM-1 at the eighth week following the injection of the cells.
  • the kinetics of the serum ESM-1 levels were also analyzed in the mice having received the transfected HEK cell expressing the glycosylated protein ESM-1 (ESM / WT).
  • the experimental results illustrated in FIG. 8 show that the tumors which have grown in mice having received the transfected HEK cells produce the protein ESM-1.
  • the amount of ESM-1 protein produced in the circulation follows the kinetics of tumor development in mice.
  • the HEK cells were transfected respectively by vectors having a DNA insert coding respectively for the wild form of ESM-1 (ESM / WT), a non-glycosylated form of ESM-1 (ESM / S137A) and a glycosylated form of ESM-1 mutated at the phenylalanine residues in positions 134 and 135 which have each been replaced by an alanine residue (ESM / 73).
  • the different transfected cells were injected subcutaneously into 5-week-old SCID mice having previously received anti-asialo GM-1 antibodies.
  • Glycosylated ESM-1 is capable of inducing tumors in mice. Neither the non-glycosylated ESM-1 protein nor the glycosylated but mutated ESM-1 protein on the phenylalanine residues at positions 134 and 135 are capable of inducing the development of tumors in SCID mice.
  • the serum level of circulating ESM-1 protein was also analyzed in the different batches of mice. The results are shown in Figure 9B.
  • FIG. 9B show that detectable levels of serum ESM-1 protein can be measured, at the eighth week following the injection of the cells, only in mice having received HEK cells expressing the glycosylated ESM-1 protein ( ESM / WT).
  • mice injected with cells expressing the non-glycosylated protein ESM-1 ESM / S137A
  • mice having received the HEK cells expressing the glycosylated and mutated protein ESM-1 HEK-ESM / F115A, F116A do not produce protein ESM-1.
  • the immuno-detection test consists of an immuno-enzymatic test of the “sandwich” type, the general characteristics of which are identical to that described by BECHARD et al. (2000).
  • the anti-ESM-1 monoclonal antibody produced by the MEP14 hybridoma line (CNCM No. l-1942) was diluted to a concentration of 5 ⁇ g / ml in 0.1 M carbonate buffer, pH 9.5, and adsorbed overnight at + 4 ° C on a 96-well plate (EIA / R.IA plate, Costar, Cambridge, MA, USA).
  • the plate was saturated for one hour at laboratory temperature with a volume of 200 ⁇ l / well of PBS buffer containing 0.1% bovine serum albumin and 5 mM EDTA, then washed twice with ELISA buffer (PBS buffer above with 0.1% Tween 20 added).
  • ELISA buffer PBS buffer above with 0.1% Tween 20 added.
  • the blood samples were diluted serially (1: 2 to 1: 128) in ELISA buffer and incubated on an ELISA plate for one hour at laboratory temperature.
  • the wells were washed three times with ELISA buffer and then incubated for 1 hour at laboratory temperature with a second monoclonal antibody directed against ESM-1, the antibody MEC15 (CNCM No. 1-2572) at the concentration of 0, 1 ⁇ g / ml in 100 ⁇ l of buffer per well.
  • a biotinylated rat monoclonal antibody directed against mouse lgG1 was added (marketed by PHARMINGEN) diluted in ELISA buffer and allowed to incubate this second antibody for one hour.
  • the streptavidin-peroxidase conjugate is revealed with the TMB substrate sold by the SIGMA company (Saint-Louis, MO, USA) in the presence of 255 ⁇ l of H 2 0 2 for 30 '.
  • the revelation reaction is stopped by the addition of a volume of 100 ⁇ l of 2N H 2 SO 4 .
  • the plate is read using a spectrophotometer (anthos labtec LP40, France) at the wavelength of 405 nanometers.
  • the concentration of the plasma or serum ESM-1 protein is calculated from the optical density measurements and expressed in nanograms per ml.
  • the concentration of circulating ESM-1 protein in the serum of different patients with bronchopulmonary cancer was measured at different stages of development, respectively in stages I, II, IIIA, IIIB and IV according to the international TNM classification which is defined below. -after:
  • T tumor size (T1: ⁇ 1 cm; T2: between 1 and 3 cm; T3:> 3 cm.
  • N lymph node (NO if not invaded; N1 if invaded).
  • M distant metastasis (MO if no meastasis; M if metastasis).
  • EXAMPLE 8 Anti-tumor activity of an ESM-1 antagonist compound of the antibody type.
  • the monoclonal antibodies MEP-08 are injected intraperitoneally at a dose of 400 ⁇ g from the second following inoculation of the HEK ESM-WT cells. The injections are repeated weekly for 12 weeks. A control antibody, MEP-14, is used under the same conditions. The mice are sacrificed when their tumor volume is greater than 6 cm 3 , (n> 8 mice in each group). The figure represents the percentage of live mice in each of the groups.
  • FIG. 10 shows that early injections of MEP-08 antibodies significantly increase mouse survival by almost 60% while MEP-14 antibodies have no effect.
  • These first results show that it is a specific action linked to the Fab fragment of the antibody directed specifically against phenylalanine at position 115 and confirm the involvement of the peptide in tumor growth. It is surprising to find that this effect on survival decreases when antibodies are administered in a delayed manner. No matter which week the injections start, antibodies can delay or prevent tumor growth. This anti-tumor effect remains more pronounced when the antibodies are used early.

Abstract

The invention concerns the use of a compound antagonist of the ESM-1 protein for making a medicine for treating cancer.

Description

UTILISATION D ' UN COMPOSE ANTAGONISTE DE LA PROTEINE ESM-1 POUR LA FABRICATION D ' UN MEDICAMENT POUR LE TRAITEMENT D ' UN CANCERUSE OF AN ANTAGONIST COMPOUND OF THE ESM-1 PROTEIN FOR THE MANUFACTURE OF A MEDICAMENT FOR THE TREATMENT OF CANCER
Domaine de l'inventionField of the invention
La présente invention se rapporte au domaine de la prévention 10 et/ou du traitement des cancers.The present invention relates to the field of cancer prevention and / or treatment.
Etat de la techniqueState of the art
En dépit des énormes investissements économiques etDespite huge economic investments and
15 humains, le cancer reste l'une des causes majeures de décès.15 humans, cancer remains one of the major causes of death.
Le cancer est fréquemment une maladie liée à des déficits dans le système de signalisation intracellulaire. Les cellules normales répondent à de nombreux signaux extracellulaires en proliférant, en se différenciant ou plus généralement en altérant leur activité métabolique.Cancer is frequently a disease linked to deficits in the intracellular signaling system. Normal cells respond to many extracellular signals by proliferating, differentiating or more generally by altering their metabolic activity.
20 De tels signaux sont reçus à la surface des cellules et convertis par un système de protéines de transduction du signal en un message reconnu par la cellule. Ce message est responsable de phénomènes de régulation cellulaire ultérieure.Such signals are received on the surface of cells and converted by a signal transduction protein system into a message recognized by the cell. This message is responsible for phenomena of subsequent cellular regulation.
La métastase est la formation d'une colonie tumoraleMetastasis is the formation of a tumor colony
25 secondaire a un site distant de la tumeur initiale. Il s'agit d'un processus multi-étapes pour lequel l'invasion tumorale est un événement précoce. Les cellules tumorales s'échappent localement à travers des barrières tissulaires, telles que la membrane basale de l'épithélium, et atteignent le stroma interstitiel, à partir duquel elles accèdent aux vaisseaux sanguins25 secondary to a site distant from the initial tumor. It is a multi-step process for which tumor invasion is an early event. Tumor cells locally escape through tissue barriers, such as the basement membrane of the epithelium, and reach the interstitial stroma, from which they access blood vessels
30 ou aux canaux lymphatiques avant dissémination ultérieure. Après avoir envahi la couche endothéliale de la paroi vasculaire, les cellules tumorales circulantes sont entraînées dans la circulation sanguine et sont stoppées dans les veinules précapillaires de l'organe cible par adhérence aux surfaces du lumen de la cellule endothéliale, ou sont30 or to the lymphatic channels before further dissemination. After invading the endothelial layer of the vascular wall, the circulating tumor cells are carried into the bloodstream and are stopped in the precapillary venules of the target organ by adhesion to the lumen surfaces of the endothelial cell, or are
35 exposées aux membranes basâtes. Les cellules tumorales sortent de la paroi vasculaire et entrent dans le parenchyme de l'organe. Enfin, la cellule tumorale, après extravasation, se multiplie dans un tissu différent de celui dont elle est originaire.35 exposed to basement membranes. The tumor cells leave the vascular wall and enter the parenchyma of the organ. Finally, the tumor cell, after extravasation, multiplies in a tissue different from that from which it originates.
Il est démontré qu'un certain nombre de cancers sont provoqués par des défauts associés à des gènes responsables de la transduction du signal. De tels gènes sont appelés encogènes. Les oncogènes peuvent conduire à une surexpression d'une ou plusieurs protéines de transduction du signal induisant une prolifération cellulaire anormale. Les signaux défectifs peuvent être liés à des mécanismes divers. Certaines thérapies anticancéreuses visent à inhiber l'expression ou la biodisponibilité de protéines oncogènes responsables de la prolifération des cellules cancéreuses, telles que les protéines de la famille des MAP kinases ou des produits de certains oncogènes tels que c-myc. La protéine ESM-1 est un polypeptide de 184 acides aminés sécrétée par les cellules endothéliales et qui a été décrite pour la première fois par LASSALLE et al. (1996). Les ARNs messagers codant pour la protéine ESM-1 sont principalement retrouvés au niveau des cellules endothéliales et des tissus pulmonaires et rénaux. L'expression du gène codant pour ESM-1 est régulée par des cytokines. Le TNF-α et L'IL-1β induisent une augmentation de l'expression du gène ESM-1 dans les cellules endothéliales de la veine ombilicale humaine, alors que l'Interféron-γ diminue son expression.A number of cancers have been shown to be caused by defects associated with genes responsible for signal transduction. Such genes are called encogenes. Oncogenes can lead to overexpression of one or more signal transducing proteins inducing abnormal cell proliferation. Defective signals can be linked to various mechanisms. Certain anticancer therapies aim to inhibit the expression or the bioavailability of oncogenic proteins responsible for the proliferation of cancer cells, such as proteins of the MAP kinase family or products of certain oncogenes such as c-myc. The ESM-1 protein is a polypeptide of 184 amino acids secreted by endothelial cells and which was described for the first time by LASSALLE et al. (1996). The messenger RNAs coding for the protein ESM-1 are mainly found in endothelial cells and in pulmonary and renal tissues. The expression of the gene encoding ESM-1 is regulated by cytokines. TNF-α and IL-1β induce an increase in the expression of the ESM-1 gene in endothelial cells of the human umbilical vein, while Interferon-γ decreases its expression.
On a retrouvé une grande quantité de protéine ESM-1 circulante chez des patients présentant un syndrome inflammatoire systémique, tel qu'un choc septique (BECHARD et al., (2000).A large amount of circulating ESM-1 protein has been found in patients with systemic inflammatory syndrome, such as septic shock (BECHARD et al., (2000).
Aujourd'hui, le traitement hospitalier du cancer a majoritairement recours à l'utilisation de radiations et/ou d'agents chimiothérapeutiques, tels que la vinblastine ou l'adriamycine. Cependant, les effets indésirables largement reconnus de tels traitements rendent ces stratégies thérapeutiques difficiles à supporter pour le patient.Today, hospital cancer treatment mainly uses the use of radiation and / or chemotherapeutic agents, such as vinblastine or adriamycin. However, the widely recognized side effects of such treatments make these therapeutic strategies difficult for the patient to bear.
La présente invention a pour objet de fournir des composés anti-cancéreux qui permettraient de surmonter les inconvénients des méthodes de traitement thérapeutique du cancer de l'état de la technique.The object of the present invention is to provide anti-cancer compounds which would make it possible to overcome the disadvantages of State of the art methods of therapeutic treatment of cancer.
SOMMAIRE DE L'INVENTIONSUMMARY OF THE INVENTION
Un premier objet de l'invention consiste en l'utilisation d'un composé antagoniste de la protéine ESM-1 pour la fabrication d'un médicament pour le traitement d'un cancer.A first object of the invention consists in the use of a compound antagonist of the ESM-1 protein for the manufacture of a medicament for the treatment of cancer.
Selon un premier aspect, un composé antagoniste de l'invention est un anticorps se liant spécifiquement à la protéine ESM-1.According to a first aspect, an antagonist compound of the invention is an antibody specifically binding to the protein ESM-1.
Selon un second aspect, un composé antagoniste mis en oeuvre dans le cadre de l'invention est un peptide d'au moins 10 acides aminés de la protéine ESM-1 modifiée et qui comprend l'enchaînement d'acides aminés Ala(134)-Ala(135). Selon un troisième aspect, un composé antagoniste de la protéine ESM-1 consiste en un oligonucléotide antisens hybridant avec l'ADNc codant pour ESM-1.According to a second aspect, an antagonist compound used in the context of the invention is a peptide of at least 10 amino acids of the modified ESM-1 protein and which comprises the chain of amino acids Ala (134) - Ala (135). According to a third aspect, an antagonist compound of the ESM-1 protein consists of an antisense oligonucleotide hybridizing with the cDNA coding for ESM-1.
Un autre objet de l'invention consiste en un composé antagoniste de la protéine ESM-1 , choisi parmi les composés antagonistes définis ci-dessus.Another object of the invention consists of an antagonist compound of the ESM-1 protein, chosen from the antagonist compounds defined above.
L'invention est également relative à une composition pharmaceutique destinée au traitement du cancer comprenant un composé antagoniste de la protéine ESM-1.The invention also relates to a pharmaceutical composition intended for the treatment of cancer comprising an antagonist compound of the protein ESM-1.
Un autre objet de l'invention consiste en une méthode de prévention d'un cancer comportant une étape au cours de laquelle on administre un composé antagoniste de la protéine ESM-1.Another subject of the invention consists of a method of preventing cancer comprising a step during which a compound antagonist of the protein ESM-1 is administered.
L'invention concerne aussi une méthode de traitement thérapeutique du cancer comportant une étape au cours de laquelle on administre à un patient un composé antagoniste de la protéine ESM-1.The invention also relates to a method of therapeutic treatment of cancer comprising a step during which a compound antagonist of the protein ESM-1 is administered to a patient.
DESCRIPTION DETAILLEE DE L'INVENTIONDETAILED DESCRIPTION OF THE INVENTION
Il a été montré pour la première fois selon l'invention que la protéine ESM-1 est sécrétée chez l'homme sous la forme d'un protéoglycane du type chondroïtine/dermatane sulfate et que la protéine ESM-1 sécrétée est capable de stimuler in vitro l'activité mitogène du facteur HGF/SF (pour « Hépatocyte growth factor/scatter factor »).It has been shown for the first time according to the invention that the protein ESM-1 is secreted in humans in the form of a proteoglycan of the chondroitin / dermatan sulfate type and that the protein Secreted ESM-1 is capable of stimulating the mitogenic activity of factor HGF / SF in vitro (for “Hepatocyte growth factor / scatter factor”).
Le HGF/SF est un facteur important dans l'apparition des dysplasies multicystiques rénales et dans l'apparition d'hyperprolifération des tubules rénaux et a été également associé au développement de carcinomes du sein, des reins et du poumon mais aussi dans le développement de mélanomes malins.HGF / SF is an important factor in the onset of renal multicystic dysplasia and in the appearance of hyperproliferation of the renal tubules and has also been associated with the development of carcinomas of the breast, kidneys and lung but also in the development of malignant melanomas.
Il a également été montré selon l'invention que des cellules épithéliales rénales humaines transfectées exprimant la protéine ESM-1 avaient un fort potentiel tumoral et provoquaient l'apparition d'un carcinome rénal in vivo chez la souris. Il a également été montré que des anticorps dirigés contre la protéine ESM-1 étaient capables d'inhiber le développement d'une tumeur rénale in vivo et qu'un peptide antagoniste de la protéine ESM-1 possédait la même activité anti-tumorale. De plus, il a été montré selon l'invention une augmentation du taux sérique de la protéine ESM-1 chez des patients présentant un carcinome broncho-pulmonaire.It has also been shown according to the invention that transfected human renal epithelial cells expressing the protein ESM-1 have a high tumor potential and cause the appearance of renal carcinoma in vivo in mice. Antibodies against the ESM-1 protein have also been shown to be capable of inhibiting the development of a renal tumor in vivo and that an antagonist peptide of the ESM-1 protein has the same anti-tumor activity. In addition, an increase in the serum ESM-1 protein level has been shown according to the invention in patients with bronchopulmonary carcinoma.
En conséquence, un premier objet de l'invention consiste en l'utilisation d'un composé antagoniste de la protéine ESM-1 pour la fabrication d'un médicament pour la prévention et/ou le traitement d'un cancer.Consequently, a first object of the invention consists in the use of an antagonist compound of the ESM-1 protein for the manufacture of a medicament for the prevention and / or treatment of cancer.
DEFINITIONS GENERALESGENERAL DEFINITIONS
Les expressions « protéine ESM-1 » ou « polypeptide ESM-1 », au sens de l'invention, englobent un polypeptide de 184 acides aminés référencé comme la séquence SEQ ID N°1 du listage de séquences, ainsi qu'un polypeptide de 165 acides aminés identique au polypeptide de séquence SEQ ID N°1 dans lequel les 19 acides aminés de l'extrémité N-terminale correspondant aux peptides signal sont absents, ce polypeptide de 165 acides aminés constituant la forme sécrétée du polypeptide de séquence SEQ ID N°1. Sont également englobés dans la définition de « protéine ESM-1 » « polypeptide ESM-1 » un glycopeptide respectivement de 184 acides aminés de séquence SEQ ID N°1 et un polypeptide de 165 acides aminés correspondant à la séquence allant de l'acide aminé en position 20 à l'acide aminé en position 184 de la séquence SEQ ID N°1 dont le résidu serine en position 137 est modifié par O-glycosylation, les formes O-glycosylées de la protéine ESM-1 étant également désignées « glycopeptides » dans la présente description. De préférence, le glycopeptide ESM-1 possède le résidu serine en position 137 qui est O-glycosylé par un motif chondroïtine/dermatane sulfate.The expressions “ESM-1 protein” or “ESM-1 polypeptide”, within the meaning of the invention, include a polypeptide of 184 amino acids referenced as the sequence SEQ ID No. 1 of the sequence listing, as well as a polypeptide of 165 amino acids identical to the polypeptide of sequence SEQ ID No. 1 in which the 19 amino acids of the N-terminal corresponding to the signal peptides are absent, this polypeptide of 165 amino acids constituting the secreted form of the polypeptide of sequence SEQ ID N 1. Also included in the definition of “ESM-1 protein” “ESM-1 polypeptide” is a glycopeptide of 184 amino acids of sequence SEQ ID No. 1 respectively and a polypeptide of 165 amino acids corresponding to the sequence ranging from the amino acid at position 20 to the amino acid at position 184 of the sequence SEQ ID No. 1 whose serine residue at position 137 is modified by O-glycosylation, the O-glycosylated forms of the protein ESM-1 also being designated "glycopeptides" in the present description. Preferably, the ESM-1 glycopeptide has the serine residue at position 137 which is O-glycosylated by a chondroitin / dermatan sulfate motif.
Par « composé antagoniste » de la protéine ESM-1 , on entend selon l'invention un composé capable de réduire significativement la biodisponiblité de la protéine ESM-1 vis-à-vis de molécules cibles sur laquelle ou lesquelles la protéine ESM-1 se fixe naturellement. Un composé antagoniste de la protéine ESM-1 peut réduire la biodisponibilité de ces protéines en réduisant la probabilité de liaison de la protéine ESM-1 aux molécules cibles de l'organisme sur lesquelles elle se fixe naturellement. Un composé antagoniste selon l'invention peut réduire la biodisponibilité de la protéine ESM-1 en inhibant ou bloquant la transcription du gène codant pour ESM-1 , en inhibant ou bloquant la traduction de l'ARN messager correspondant, en altérant la maturation intracellulaire de la protéine ESM-1 , par exemple en affectant le processus enzymatique conduisant à sa glycosylation, ou encore en inhibant ou en bloquant la sécrétion de la protéine ESM-1 mature.By “antagonist compound” of the protein ESM-1, is meant according to the invention a compound capable of significantly reducing the bioavailability of the protein ESM-1 with respect to target molecules on which which the protein ESM-1 is naturally fixes. An ESM-1 protein antagonist compound can reduce the bioavailability of these proteins by reducing the likelihood of binding of the ESM-1 protein to target molecules in the body to which it naturally binds. An antagonist compound according to the invention can reduce the bioavailability of the protein ESM-1 by inhibiting or blocking the transcription of the gene coding for ESM-1, by inhibiting or blocking the translation of the corresponding messenger RNA, by altering the intracellular processing of the ESM-1 protein, for example by affecting the enzymatic process leading to its glycosylation, or by inhibiting or blocking the secretion of the mature ESM-1 protein.
Un premier objet de l'invention consiste en l'utilisation d'un composé antagoniste de la protéine ESM-1 pour la fabrication d'un médicament pour le traitement d'un cancer.A first object of the invention consists in the use of a compound antagonist of the ESM-1 protein for the manufacture of a medicament for the treatment of cancer.
Un composé antagoniste de la protéine ESM-1 peut être de toute nature, polypeptide, saccharidique, ou encore tout composé organique ou minérale permettant de réduire la biodisponibilité de la protéine ESM-1 vis-à-vis des molécules cibles sur laquelle cette protéine se fixe.An ESM-1 protein antagonist compound may be of any kind, polypeptide, saccharide, or any organic or inorganic compound making it possible to reduce the bioavailability of the ESM-1 protein with respect to the target molecules on which this protein is fixed.
COMPOSES ANTAGONISTES DE LA PROTEINE ESM-1 DU TYPE ANTICORPSANTAGONIST COMPOUNDS OF THE ESM-1 PROTEIN OF THE ANTIBODY TYPE
Une première famille de composés antagonistes de ESM-1 préférée selon l'invention est constituée par des anticorps se liant spécifiquement à la protéine ESM-1. Il est montré selon l'invention que des anticorps dirigés spécifiquement contre la protéine ESM-1 sont capables d'inhiber ou de bloquer le pouvoir tumorigène de cette protéine. Des anticorps anti-ESM- 1 constituent donc des composés antagonistes d'une grande valeur thérapeutique.A first family of ESM-1 antagonist compounds preferred according to the invention consists of antibodies specifically binding to the ESM-1 protein. It is shown according to the invention that antibodies directed specifically against the protein ESM-1 are capable of inhibiting or blocking the tumorigenicity of this protein. Anti-ESM-1 antibodies therefore constitute antagonistic compounds of great therapeutic value.
Par « anticorps » au sens de l'invention, on entend notamment des anticorps polyclonaux ou monoclonaux ou des fragments (par exemple les fragments Fab ou F(ab)'2 ou encore tout polypeptide comprenant un domaine de l'anticorps initial reconnaissant la protéine ESM-1.By “antibody” within the meaning of the invention is meant in particular polyclonal or monoclonal antibodies or fragments (for example the Fab or F (ab) ' 2 fragments or any polypeptide comprising a domain of the initial antibody recognizing the protein ESM-1.
Des anticorps monoclonaux peuvent être préparés à partir d'hybridome selon la technique décrite par KOHLER et MIELSTEIN (1975).Monoclonal antibodies can be prepared from a hybridoma using the technique described by KOHLER and MIELSTEIN (1975).
Il peut s'agir aussi des anticorps dirigés contre. ESM-1 ou un fragment de cette protéine produits par la technique du trioma ou encore la technique d'hybridome décrite par KOZBOR et al. (1983).It can also be antibodies directed against. ESM-1 or a fragment of this protein produced by the trioma technique or also the hybridoma technique described by KOZBOR et al. (1983).
Il peut s'agir également de fragments d'anticorps simple chaîne Fv (ScFv) tels que ceux décrits dans le brevet US N04,9476,778 ou encore par MARTINEAU et al. (1998). Des anticorps anti-ESM-1 selon l'invention comprennent également des fragments d'anticorps obtenus à l'aide de banques de phages telles que décrites par RIDDER et al. (1995) ou encore des anticorps humanisés tels que décrits par REINMANN et al. (1997) ou encore par LEGER OJ, et al., 1997. II peut s'agir également des anticorps anti-ESM-1 produits selon les techniques décrites par BECHARD et al. (2000). Les anticorps décrits par BECHARD et al. (2000) sont des anticorps monoclonaux sécrétés par des lignées d'hybridome préparées à partir de cellules spléniques de souris préalablement immunisées contre le fragment C-terminal de poids moléculaire de 14 kD de ESM-1 qui a été produit dans Escherichia coli, c'est-à-dire un fragment non glycosylé de la protéine ESM-1. Par cartographie d'épitopes, BECHARD et al. (2000) ont pu classer les anticorps monoclonaux produits par les différentes lignées d'hybridome selon la région de la protéine ESM-1 reconnue par ceux-ci. Une première famille d'anticorps préférée selon l'invention qui constitue des composés antagonistes de la protéine ESM-1 sont les anticorps monoclonaux reconnaissant spécifiquement la région allant du résidu praline en position 79 jusqu'au résidu cystéine en position 99 de la séquence SEQ ID N°1 , cette région représentant le déterminant antigénique D1. Il s'agit de préférence des anticorps monoclonaux produits par la lignée d'hybridome déposée le 19 Novembre 1997 auprès de la Collection Nationale de Cultures de Micro-organismes de l'Institut Pasteur (CNCM) sous le n° d'accès N°l-1944, encore appelé anticorps MEP21.They can also be fragments of single chain Fv antibody (ScFv) such as those described in US Patent No. 0 4,9476,778 or by MARTINEAU et al. (1998). Anti-ESM-1 antibodies according to the invention also comprise fragments of antibodies obtained using phage banks as described by RIDDER et al. (1995) or humanized antibodies as described by REINMANN et al. (1997) or also by LEGER OJ, et al., 1997. They may also be anti-ESM-1 antibodies produced according to the techniques described by BECHARD et al. (2000). The antibodies described by BECHARD et al. (2000) are monoclonal antibodies secreted by hybridoma lines prepared from spleen cells from mice previously immunized against the 14 kD C-terminal molecular weight fragment of ESM-1 which was produced in Escherichia coli, c ' that is to say an unglycosylated fragment of the protein ESM-1. By epitope mapping, BECHARD et al. (2000) were able to classify the monoclonal antibodies produced by the various hybridoma lines according to the region of the protein ESM-1 recognized by them. A first preferred family of antibodies according to the invention which constitute antagonist compounds of the ESM-1 protein are the monoclonal antibodies specifically recognizing the region going from the praline residue in position 79 to the cysteine residue in position 99 of the sequence SEQ ID No. 1, this region representing the antigenic determinant D1. They are preferably monoclonal antibodies produced by the hybridoma line deposited on November 19, 1997 with the National Collection of Cultures of Microorganisms of the Pasteur Institute (CNCM) under the access number N ° l -1944, also called MEP21 antibody.
D'autres anticorps monoclonaux préférés sont eux se liant spécifiquement à la partie de la protéine ESM-1 comprise entre le résidu glycine en position 159 et le résidu arginine en position 184 de la séquence SEQ ID N°1 qui est la région constituant le déterminant antigénique D3. Des anticorps monoclonaux préférés spécifiques du déterminant antigénique D3 peuvent être obtenus à partir de la lignée d'hybridome 1-1943 (MEP19), déposée le 19 Novembre 1997 auprès de la Collection Nationale de Cultures des Micro-organismes de l'Institut Pasteur (CNCM). D'autres anticorps monoclonaux préférés selon l'invention sont les anticorps monoclonaux se liant spécifiquement avec la région comprise entre le résidu serine en position 119 et le résidu valine en position 139 de la protéine ESM-1 de séquence SEQ ID N°1 , cette région étant définie comme le déterminant antigénique D2 de la protéine ESM- 1. Les anticorps monoclonaux préférés se liant spécifiquement au déterminant antigénique D2 de ESM-1 peuvent être obtenus à partir de la lignée d'hybridome MEP08 déposée le 19 Novembre 1997 auprès de la Collection Nationale de Cultures de Micro-organismes de l'Institut Pasteur (CNCM) sous le n° d'accès N° 1-1941. D'autres anticorps monoclonaux d'intérêt constituant des composés antagonistes de la protéine ESM-1 , au sens de l'invention, sont les anticorps monoclonaux dirigés spécifiquement contre la partie N- terminale de la protéine ESM-1. Les anticorps monoclonaux préférés dirigés contre la partie N-terminale de la protéine ESM-1 peuvent être obtenus à partir de la lignée d'hybridome MEC15 déposée à la Collection Nationale de Cultures des Micro-organismes de l'INSTITUT PASTEUR (CNCM) le 17 Octobre 2000 sous le numéro d'accès I-2572.Other preferred monoclonal antibodies are they which specifically bind to the part of the ESM-1 protein between the glycine residue at position 159 and the arginine residue at position 184 of the sequence SEQ ID No. 1 which is the region constituting the determinant D3 antigen. Preferred monoclonal antibodies specific for the antigenic determinant D3 can be obtained from the hybridoma line 1-1943 (MEP19), deposited on November 19, 1997 with the National Collection of Cultures of Microorganisms of the Institut Pasteur (CNCM ). Other preferred monoclonal antibodies according to the invention are the monoclonal antibodies binding specifically with the region between the serine residue at position 119 and the valine residue at position 139 of the protein ESM-1 of sequence SEQ ID No. 1, this region being defined as the antigenic determinant D2 of the protein ESM-1. The preferred monoclonal antibodies specifically binding to the antigenic determinant D2 of ESM-1 can be obtained from the hybridoma line MEP08 deposited on November 19, 1997 with the National Collection of Cultures of Microorganisms of the Institut Pasteur (CNCM) under the access number N ° 1-1941. Other monoclonal antibodies of interest constituting antagonist compounds of the ESM-1 protein, within the meaning of the invention, are the monoclonal antibodies directed specifically against the N-terminal part of the ESM-1 protein. The preferred monoclonal antibodies directed against the N-terminal part of the ESM-1 protein can be obtained from the hybridoma line MEC15 deposited in the Collection National Culture of Microorganisms of the INSTITUT PASTEUR (CNCM) on October 17, 2000 under the access number I-2572.
Selon un aspect tout à fait préféré, les anticorps anti-ESM1 possédant les meilleures activités antagonistes vis-à-vis de ESM-1 sont choisis parmi les anticorps reconnaissant spécifiquement des epitopes localisés dans la région encadrant le résidu phénylalanine en position 115. Il s'agit notamment des anticorps se liant spécifiquement avec la région comprise entre le résidu serine en position 119 et le résidu valine en position 139 de la protéine ESM-1 de séquence SEQ ID N°1 , tel que l'anticorps monoclonal MEP08 décrit ci-dessus.According to an entirely preferred aspect, the anti-ESM1 antibodies having the best antagonistic activities vis-à-vis ESM-1 are chosen from antibodies specifically recognizing epitopes located in the region surrounding the phenylalanine residue at position 115. It s acts in particular antibodies which bind specifically with the region between the serine residue at position 119 and the valine residue at position 139 of the protein ESM-1 of sequence SEQ ID No. 1, such as the monoclonal antibody MEP08 described above above.
Il a été montré selon l'invention que l'anticorps monoclonal MEP08 était capable d'inhiber l'activité pro-tumorale de la protéine ESM- 1 sur la formation de tumeurs provoquées par la prolifération de cellules humaines d'origine rénale chez la souris.It has been shown according to the invention that the monoclonal antibody MEP08 was capable of inhibiting the pro-tumor activity of the protein ESM-1 on the formation of tumors caused by the proliferation of human cells of renal origin in mice .
ROLYPEPTIDES ANTAGONISTES DE LA PROTEINE ESM-1ESM-1 PROTEIN ANTAGONIST ROLYPEPTIDES
Il a été montré selon l'invention que la région recouvrant le déterminant antigénique D2 de la protéine ESM-1 était importante pour l'activité pro-tumorale de la protéine ESM-1.It has been shown according to the invention that the region covering the antigenic determinant D2 of the protein ESM-1 is important for the pro-tumor activity of the protein ESM-1.
En particulier, le demandeur a synthétisé un polypeptide dérivé de la protéine ESM-1 dans lequel les résidus phénylalanine en positions 134 et 135 de la séquence SEQ ID N°1 , c'est-à-dire les résidus en positions 115 et 116 de la protéine ESM-1 sécrétée, ont été remplacés par deux résidus alanine. Le demandeur a montré que ce polypeptide modifié était incapable d'induire des tumeurs chez la souris. Un tel polypeptide modifié peut donc entrer en compétition avec la protéine ESM-1 produite à haut niveau chez des patients cancéreux pour son action potentialisatrice avec des facteurs de croissance comme le HGF/SF ou encore les facteurs de croissance FGF-2 et FGF-7.In particular, the applicant has synthesized a polypeptide derived from the ESM-1 protein in which the phenylalanine residues in positions 134 and 135 of the sequence SEQ ID No. 1, that is to say the residues in positions 115 and 116 of the secreted protein ESM-1, have been replaced by two alanine residues. The applicant has shown that this modified polypeptide is incapable of inducing tumors in mice. Such a modified polypeptide can therefore compete with the protein ESM-1 produced at high level in cancer patients for its potentiating action with growth factors such as HGF / SF or even growth factors FGF-2 and FGF-7 .
Les composés antagonistes de la protéine ESM-1 englobent des polypeptides ayant une longueur d'au moins 10 acides aminés consécutifs de la séquence SEQ ID N°1 , qui comprennent une séquence d'acides aminés allant de l'acide aminé en position 119 jusqu'à l'acide aminé en position 139 de la séquence SEQ ID N°1, un tel polypeptide antagoniste de ESM-1 comprenant au moins une substitution d'un acide aminé, par rapport à la séquence correspondante de la protéine ESM-1.ESM-1 protein antagonist compounds include polypeptides of at least 10 consecutive amino acids in sequence SEQ ID NO: 1, which include an amino acid sequence ranging from amino acid at position 119 to with the amino acid in position 139 of the sequence SEQ ID No. 1, such a polypeptide ESM-1 antagonist comprising at least one substitution of an amino acid, with respect to the corresponding sequence of the ESM-1 protein.
De préférence, un polypeptide antagoniste de la protéine ESM- 1 tel que défini ci-dessus possède au plus 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 30, 35, 40, 45 ou 50 acides aminés consécutifs de la séquence SEQ ID N°1 et au moins une substitution d'acides aminés, par rapport à la séquence SEQ ID n°1 .Preferably, an ESM-1 protein antagonist polypeptide as defined above has at most 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 , 30, 35, 40, 45 or 50 consecutive amino acids of the sequence SEQ ID No 1 and at least one substitution of amino acids, compared to the sequence SEQ ID No 1.
Un polypeptide antagoniste de la protéine ESM-1 , tel que défini ci-dessus, comprend au plus 1 , 2, 3, 4, 5, 6, 7, 8, 9 ou 10 substitutions d'un acide aminé, par rapport à la séquence SEQ ID N°1 , le nombre de substitutions d'acides aminés étant adapté en fonction de la longueur du polypeptide, étant entendu que le nombre de substitutions d'acides aminés par rapport à la séquence SEQ ID N°1 dans un polypeptide antagoniste selon l'invention est d'au plus 25% des acides aminés compris dans la séquence de ce polypeptide antagoniste, de préférence au plus 20%, 15% et de manière préférée d'au plus 10% du nombre des acides aminés compris dans la séquence du polypeptide antagoniste de ESM-1.An ESM-1 protein antagonist polypeptide, as defined above, comprises at most 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid substitutions, relative to the sequence SEQ ID No 1, the number of amino acid substitutions being adapted as a function of the length of the polypeptide, it being understood that the number of amino acid substitutions relative to the sequence SEQ ID No 1 in an antagonistic polypeptide according to the invention is at most 25% of the amino acids included in the sequence of this antagonistic polypeptide, preferably at most 20%, 15% and preferably at most 10% of the number of amino acids included in the ESM-1 antagonist polypeptide sequence.
De manière préférée, une substitution d'acides aminés, par rapport à la séquence SEQ ID N°1 , dans un polypeptide antagoniste selon l'invention est une substitution dite « non conservative ». Par substitution « non conservative », on entend la substitution d'un résidu d'acide aminé par un acide aminé d'une classe distincte.Preferably, an amino acid substitution, with respect to the sequence SEQ ID No. 1, in an antagonistic polypeptide according to the invention is a so-called “non-conservative” substitution. By "non-conservative" substitution is meant the substitution of an amino acid residue with an amino acid of a distinct class.
Les acides aminés sont classiquement rangés selon les classes suivantes:Amino acids are classically classified according to the following classes:
- acides aminés non polaires (hydrophobes): alanine, leucine, isoleucine, valine, praline, phénylalanine, tryptophane et méthionine;- non-polar (hydrophobic) amino acids: alanine, leucine, isoleucine, valine, praline, phenylalanine, tryptophan and methionine;
- acides aminés contenant des cycles aromatiques: phénylalanine, tryptophane et tyrosine; - acides aminés neutres polaires: glycine, serine, thréonine, cystéine, tyrosine, asparagine et glutamine;- amino acids containing aromatic rings: phenylalanine, tryptophan and tyrosine; - polar neutral amino acids: glycine, serine, threonine, cysteine, tyrosine, asparagine and glutamine;
- acides aminés chargés positivement (basiques) : arginine, lysine et histidine);- positively charged (basic) amino acids: arginine, lysine and histidine);
- acides aminés chargés négativement (acides): acide aspartique et acide glutamique. Un type de substitution d'acides aminés préféré pour la préparation d'un polypeptide antagoniste de la protéine ESM-1 selon l'invention est la substitution d'un acide aminé contenant un cycle aromatique par un acide aminé ne contenant pas de cycle aromatique. De manière tout à fait préférée, un polypeptide antagoniste de la protéine ESM-1 selon l'invention comprend une substitution des résidus phénylalanines en positions 134 et 135 de la SEQ ID N°1 par deux résidus d'acides aminés, identiques ou différents, ne contenant pas de cycle aromatique . Un tel polypeptide antagoniste de la protéine ESM-1 préféré est un polypeptide d'au moins 10 acides aminés consécutifs de la séquence SEQ ID N°1 , telle que définie ci-dessus, dans lequel les résidus phénylalanine en positions 134 et 135 ont été remplacés par deux résidus alanine. Selon un premier aspect, un polypeptide antagoniste de la protéine ESM-1 selon l'invention peut être préparé par les techniques classiques de synthèse chimique, indifféremment en solution homogène ou en phase solide.- negatively charged amino acids (acids): aspartic acid and glutamic acid. A preferred type of amino acid substitution for the preparation of an ESM-1 protein antagonist polypeptide according to the invention is the substitution of an amino acid containing an aromatic ring with an amino acid not containing an aromatic ring. Most preferably, an antagonist polypeptide of the ESM-1 protein according to the invention comprises a substitution of the phenylalanine residues in positions 134 and 135 of SEQ ID No. 1 with two amino acid residues, identical or different, not containing an aromatic cycle. Such a preferred ESM-1 protein antagonist polypeptide is a polypeptide of at least 10 consecutive amino acids of the sequence SEQ ID No. 1, as defined above, in which the phenylalanine residues at positions 134 and 135 have been replaced by two alanine residues. According to a first aspect, an antagonist polypeptide of the ESM-1 protein according to the invention can be prepared by conventional techniques of chemical synthesis, either in homogeneous solution or in solid phase.
A titre illustratif, un polypeptide antagoniste de la protéine ESM- 1 peut être préparé par la technique en solution homogène décrite par HOUBEN WEIL (1974) ou encore la technique de synthèse en phase solide décrite par MERRIFIELD (1965a; 1965b) et MERRIFIELD 1965b.As an illustration, an ESM-1 protein antagonist polypeptide can be prepared by the homogeneous solution technique described by HOUBEN WEIL (1974) or the solid phase synthesis technique described by MERRIFIELD (1965a; 1965b) and MERRIFIELD 1965b.
Un polypeptide antagoniste de la protéine ESM-1 selon l'invention peut être aussi préparé par recombinaison génétique. Pour produire un polypeptide antagoniste de la protéine ESM-1 tel que défini ci-dessus, on peut mettre en oeuvre un procédé comprenant les étapes de: a) insérer un acide nucléique codant pour le polypeptide antagoniste de la protéine ESM-1 dans un vecteur d'expression approprié; b) cultiver, dans un milieu de culture approprié, une cellule hôte préalablement transformée ou transfectée avec le vecteur d'expression recombinante de l'étape a); c) récupérer le milieu de culture conditionné ou lyser la cellule hôte, par exemple par sonication ou par choc osmotique; d) séparer et purifier à partir dudit milieu de culture ou encore à partir des lysats cellulaires obtenus à l'étape c), ledit polypeptide antagoniste; e) le cas échéant, caractériser le polypeptide antagoniste recombinant ainsi produit.An ESM-1 protein antagonist polypeptide according to the invention can also be prepared by genetic recombination. To produce an ESM-1 protein antagonist polypeptide as defined above, a method can be implemented comprising the steps of: a) inserting a nucleic acid coding for the ESM-1 protein antagonist polypeptide in a vector appropriate expression; b) cultivating, in an appropriate culture medium, a host cell previously transformed or transfected with the recombinant expression vector of step a); c) recovering the conditioned culture medium or lysing the host cell, for example by sonication or by osmotic shock; d) separating and purifying from said culture medium or also from the cell lysates obtained in step c), said antagonist polypeptide; e) where appropriate, characterize the recombinant antagonist polypeptide thus produced.
Les polypeptides antagonistes selon l'invention peuvent être caractérisés par fixation sur une colonne de chromatographie d'immunoaffinité sur laquelle les anticorps dirigés contre ce polypeptide ou contre un fragment de ce dernier ont été préalablement immobilisés. Selon un autre aspect, un polypeptide antagoniste de ESM-1 peut-être purifié par passage sur une série appropriée de colonnes de chromatographie, selon les méthodes connues de l'homme de l'art est décrit par exemple dans AUSUBEL F. et al. (1989).The antagonistic polypeptides according to the invention can be characterized by fixation on an immunoaffinity chromatography column on which the antibodies directed against this polypeptide or against a fragment of the latter have been immobilized beforehand. In another aspect, an ESM-1 antagonist polypeptide can be purified by passage through an appropriate series of chromatography columns, according to methods known to those skilled in the art is described for example in AUSUBEL F. et al. (1989).
COMPOSES ANTAGONISTES DE LA PROTEINE ESM-1 DU TYPE OLIGONUCLEOTIDE ANTISENS.ANTAGONIST COMPOUNDS OF THE ESM-1 PROTEIN OF THE ANTISENSE OLIGONUCLEOTIDE TYPE.
Une autre famille préférée de composés antagonistes de la protéine ESM-1 visant à réduire la biodisponibilité de la protéine ESM-1 sécrétée chez des patients à risque ou chez des patients ayant déjà développé des tumeurs sont des composés capables d'inhiber ou de bloquer l'expression du gène codant pour ESM-1 chez l'homme.Another preferred family of ESM-1 antagonist compounds aimed at reducing the bioavailability of the secreted ESM-1 protein in patients at risk or in patients who have already developed tumors are compounds capable of inhibiting or blocking expression of the gene encoding ESM-1 in humans.
De tels composés antagonistes de la protéine ESM-1 peuvent être des polynucléotides antisens. Les composés antagonistes de la protéine ESM-1 selon l'invention englobent ainsi un polynucléotide antisens capable de s'hybrider spécifiquement à une région déterminée du gène codant pour la protéine ESM-1 et capable d'inhiber ou de bloquer sa transcription et/ou sa traduction. La séquence du gène humain ESM-1 est référencée sous le numéro d'accès AJ401 1091 et AJ401 1092 dans la base de données Genbank.Such ESM-1 protein antagonist compounds can be antisense polynucleotides. The antagonist compounds of the ESM-1 protein according to the invention thus include an antisense polynucleotide capable of hybridizing specifically to a determined region of the gene coding for the protein ESM-1 and capable of inhibiting or blocking its transcription and / or its translation. The sequence of the human ESM-1 gene is referenced under the access number AJ401 1091 and AJ401 1092 in the Genbank database.
De préférence, un polynucléotide antisens selon l'invention comprend une séquence complémentaire d'une séquence localisée dans la région de l'extrémité 5' de l'ADN du gène ESM-1 , et de manière tout à fait préférée la proximité du codon d'initiation de la traduction (ATG) du gène ESM-1.Preferably, an antisense polynucleotide according to the invention comprises a sequence complementary to a sequence localized in the region of the 5 ′ end of the DNA of the ESM-1 gene, and so completely preferred is the proximity of the translation initiation codon (ATG) of the ESM-1 gene.
Selon un second mode de réalisation préférentiel, un polynucléotide antisens selon l'invention comprend une séquence complémentaire à l'une des séquences localisées au niveau des jonctions exon/intron du gène ESM-1 et de manière préférée des séquences correspondant à un site d'épissage.According to a second preferred embodiment, an antisense polynucleotide according to the invention comprises a sequence complementary to one of the sequences located at the exon / intron junctions of the ESM-1 gene and preferably sequences corresponding to a site of splicing.
Un polynucléotide antisens préféré selon l'invention comprend au moins 15 nucléotides consécutifs de l'ADNc codant pour ESM-1 ayant la séquence nucléotidique SEQ ID N°2.A preferred antisense polynucleotide according to the invention comprises at least 15 consecutive nucleotides of the cDNA coding for ESM-1 having the nucleotide sequence SEQ ID No. 2.
Aux fins de la présente invention, un premier polynucléotide est considéré comme étant « complémentaire » d'un second polynucléotide lorsque chaque base du premier nucléotide est appariée à la base complémentaire du second polynucléotide dont l'orientation est inversée. Les bases complémentaires sont A et T (ou A et U), et C et G.For the purposes of the present invention, a first polynucleotide is considered to be “complementary” to a second polynucleotide when each base of the first nucleotide is paired with the base complementary to the second polynucleotide whose orientation is reversed. The complementary bases are A and T (or A and U), and C and G.
De manière générale, un polynucléotide antisens selon l'invention possède au moins 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 300, 400, 500, 1000 ou 2000 nucléotides consécutifs de l'ADNc de ESM- 1 de séquence SEQ ID N°2. A titre illustratif, un polynucléotide antisens préféré selon l'invention consiste en un acide nucléique de séquence complémentaire à l'acide nucléique de l'ADNc de ESM-1 de séquence SEQ ID N°2.In general, an antisense polynucleotide according to the invention has at least 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 300, 400, 500, 1000 or 2000 consecutive nucleotides of the cDNA of ESM-1 of sequence SEQ ID No. 2. By way of illustration, a preferred antisense polynucleotide according to the invention consists of a nucleic acid of sequence complementary to the nucleic acid of the cDNA of ESM-1 of sequence SEQ ID No. 2.
Un polynucléotide antisens constituant un composé antagoniste de la protéine ESM-1 selon l'invention peut être préparé par toute méthode adaptée bien connue de l'homme du métier, y compris par clonage et action d'enzyme de restriction ou encore par synthèse chimique directe selon des techniques telles que la méthode au phosphodiester de NARANG et al. (1979) ou de BROWN et al. (1979), la méthode au diéthylphosphoramidites de BEAUCAGE et al. (1980) ou encore la technique sur support solide décrite dans le brevet européen n°EP-0 707 592.An antisense polynucleotide constituting an antagonist compound of the ESM-1 protein according to the invention can be prepared by any suitable method well known to those skilled in the art, including by cloning and restriction enzyme action or by direct chemical synthesis. according to techniques such as the phosphodiester method of NARANG et al. (1979) or BROWN et al. (1979), the diethylphosphoramidite method of BEAUCAGE et al. (1980) or the solid support technique described in European patent No. EP-0 707 592.
De manière générale, les polynucléotides antisens doivent avoir une longueur et une température de fusion suffisante pour permettre la formation d'un hybride duplex intracellulaire ayant une stabilité suffisante pour inhiber l'expression de l'ARNm de ESM-1. Des stratégies pour construire les polynucléotides antisens sont notamment décrites par GREEN et al. (1986) et IZANT et WEINTRAUB (1984).In general, the antisense polynucleotides must have a sufficient length and melting temperature to allow the formation of an intracellular duplex hybrid having sufficient stability to inhibit expression of the ESM-1 mRNA. Strategies for constructing the antisense polynucleotides are notably described by GREEN et al. (1986) and IZANT and WEINTRAUB (1984).
Des méthodes de construction de polynucléotides antisens sont également décrites par ROSSI et al (1991 ) ainsi que dans les demandes PCT N°WO 947/23.026, WO 95/04141 , WO 92/L18.522 et dans la demande de brevet européen n°EP 0 572 287.Methods for constructing antisense polynucleotides are also described by ROSSI et al (1991) as well as in PCT applications No. WO 947 / 23.026, WO 95/04141, WO 92 / L18.522 and in European patent application No. EP 0 572 287.
D'autres méthodes de mise en oeuvre de polynucléotides antisens sont par exemple celles décrites par SCZAKIEL et al. (1995) ou encore celles décrites dans la demande PCT N°WO 95/24.223. L'homme du métier peut avantageusement se référer aux méthodes de production et d'utilisation de polynucléotides antisens inhibant ou bloquant l'expression de gènes associés au développement de cancers, telles que les techniques décrites dans le brevet US N°5,582,986 qui divulgue des oligonucléotides antisens pour l'inhibition du gène ras, la technique décrite par HOLT et al. (1988) qui décrit des oligonucléotides antisens s'hybridant spécifiquement avec les ARNs messagers l'oncogène c-myb ou encore la technique décrite par WICKSTRON et al. (1988) qui décrit des oligonucléotides antisens hybridant spécifiquement avec l'ARN messager du gène c-myc. D'autres techniques d'utilisation de polynucléotides antisens utilisables par l'homme du métier sont celles de SALE et al. (1995) ainsi que celle de GAO et al. (1996).Other methods of implementing antisense polynucleotides are for example those described by SCZAKIEL et al. (1995) or those described in PCT application No. WO 95 / 24.223. Those skilled in the art can advantageously refer to the methods of production and use of antisense polynucleotides inhibiting or blocking the expression of genes associated with the development of cancers, such as the techniques described in US Patent No. 5,582,986 which discloses oligonucleotides antisense for the inhibition of the ras gene, the technique described by HOLT et al. (1988) which describes antisense oligonucleotides which hybridize specifically with messenger RNAs the c-myb oncogene or the technique described by WICKSTRON et al. (1988) which describes antisense oligonucleotides which hybridize specifically with the messenger RNA of the c-myc gene. Other techniques for using antisense polynucleotides which can be used by those skilled in the art are those of SALE et al. (1995) as well as that of GAO et al. (1996).
PROCEDE DE SELECTION D'UN COMPOSE ANTAGONISTE DE LA PROTEINE ESM-1METHOD FOR SELECTING AN ANTAGONIST COMPOUND OF THE ESM-1 PROTEIN
Un composé antagoniste de la protéine ESM-1 selon l'invention peut être sélectionné par l'homme du métier pour sa capacité à inhiber le développement d'une tumeur induite par la protéine ESM-1 in vivo. Selon un premier aspect, un procédé de sélection d'un composé antagoniste de la protéine ESM-1 comprend les étapes suivantes: a) injecter à un animal des cellules capables de former des tumeurs en présence de la protéine ESM-1 , lesdites cellules étant transfectees ou transformées par un acide nucléique capable d'exprimer la protéine ESM-1 in vivo; b) administrer à cet animal un composé candidat antagoniste de la protéine ESM-1 ; c) comparer la formation de tumeurs chez un premier animal tel qu'obtenu à la fin de l'étape b) et chez un second animal tel qu'obtenu à la fin de l'étape a); et d) sélectionner le composé candidat capable d'inhiber ou de bloquer la formation de tumeurs chez le premier animal. De préférence, l'animal mis en oeuvre dans le procédé de sélection ci-dessus est un mammifère non humain, avantageusement un rongeur, et de manière tout à fait préférée un rat, un cobaye, un cochon d'Inde ou une souris.An ESM-1 protein antagonist compound according to the invention can be selected by a person skilled in the art for its ability to inhibit the development of a tumor induced by the ESM-1 protein in vivo. According to a first aspect, a method for selecting an antagonist compound of the ESM-1 protein comprises the following steps: a) injecting into an animal cells capable of forming tumors in the presence of the ESM-1 protein, said cells being transfected or transformed with a nucleic acid capable of expressing the ESM-1 protein in vivo; b) administering to this animal a candidate antagonist compound of the ESM-1 protein; c) comparing the formation of tumors in a first animal as obtained at the end of step b) and in a second animal as obtained at the end of step a); and d) selecting the candidate compound capable of inhibiting or blocking the formation of tumors in the first animal. Preferably, the animal used in the above selection process is a non-human mammal, advantageously a rodent, and very preferably a rat, a guinea pig, a guinea pig or a mouse.
Dans un mode de réalisation particulier du procédé, celui-ci comprend une étape e) consistant à sacrifier le premier et le second animal.In a particular embodiment of the method, it comprises a step e) consisting in sacrificing the first and the second animal.
Avantageusement, la lignée cellulaire capable de former des tumeurs chez l'animal en présence de la protéine ESM-1 est la lignéeAdvantageously, the cell line capable of forming tumors in animals in the presence of the protein ESM-1 is the line
HEK 293 (ATCC N°CRL 1573). Selon un autre aspect, un composé antagoniste de la protéineHEK 293 (ATCC N ° CRL 1573). In another aspect, a protein antagonist compound
ESM-1 selon l'invention peut être sélectionné selon un procédé permettant de mettre en évidence la fixation d'un composé candidat sur la protéine ESM-1. Un tel procédé de sélection d'un composé candidat antagoniste de la protéine ESM-1 comprend les étapes suivantes: a) fournir un polypeptide consistant en la protéine ESM-1 ou un fragment peptidique de cette protéine; b) mettre en contact ledit polypeptide avec le composé candidat à tester; c) détecter les complexes formés entre ledit polypeptide et le composé candidat; d) sélectionner les composés candidats se fixant sur le polypeptide constitué de la protéine ESM-1 ou d'un fragment de cette protéine.ESM-1 according to the invention can be selected according to a method making it possible to demonstrate the binding of a candidate compound to the protein ESM-1. Such a method for selecting a candidate antagonist compound for the ESM-1 protein comprises the following steps: a) providing a polypeptide consisting of the ESM-1 protein or a peptide fragment of this protein; b) bringing said polypeptide into contact with the candidate compound to be tested; c) detecting the complexes formed between said polypeptide and the candidate compound; d) selecting the candidate compounds which bind to the polypeptide consisting of the ESM-1 protein or of a fragment of this protein.
Par « fragment » de la protéine ESM-1 , on entend un polypeptide comprenant au moins 20, de préférence au moins 30, 35, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140 ou 150 acides aminés consécutifs du polypeptide ESM-1 de séquence SEQ ID N°1 et comprenant la séquence allant du résidu praline en position 133 jusqu'au résidu valine en position 138 de la SEQ ID N°1. L'invention est également relative à une trousse ou kit pour la sélection d'un composé candidat antagoniste de la protéine ESM-1 , cette trousse ou kit comprenant: a) une préparation purifiée d'un polypeptide constitué de la protéine ESM-1 ou d'un fragment de cette protéine; b) le cas échéant, des moyens de détection d'un complexe formé entre le polypeptide et le composé candidat à tester.By “fragment” of the ESM-1 protein is meant a polypeptide comprising at least 20, preferably at least 30, 35, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140 or 150 consecutive amino acids of the ESM-1 polypeptide of sequence SEQ ID No. 1 and comprising the sequence going from the praline residue in position 133 to the valine residue in position 138 of SEQ ID N ° 1. The invention also relates to a kit or kit for the selection of a candidate antagonist compound of the ESM-1 protein, this kit or kit comprising: a) a purified preparation of a polypeptide consisting of the ESM-1 protein or a fragment of this protein; b) where appropriate, means for detecting a complex formed between the polypeptide and the candidate compound to be tested.
Le procédé de détection d'un complexe formé entre le polypeptide dérivé de la protéine ESM-1 et le composé candidat peut- être réalisé par diverses techniques, telles que par microdialyse couplée à une méthode HPLC comme décrit par WANG et al. (1997) ou l'électrophorèse capillaire d'affinité comme décrit par BOUSH et al. (1997).The method for detecting a complex formed between the polypeptide derived from the ESM-1 protein and the candidate compound can be carried out by various techniques, such as by microdialysis coupled to an HPLC method as described by WANG et al. (1997) or capillary affinity electrophoresis as described by BOUSH et al. (1997).
Un composé candidat peut être de toute nature, et notamment le produit final d'un procédé de chimie combinatoire.A candidate compound can be of any kind, and in particular the final product of a combinatorial chemistry process.
A. COMPOSES CANDIDATS OBTENUS A PARTIR DE BANQUES DE PEPTIDESA. CANDIDATE COMPOUNDS OBTAINED FROM PEPTIDE BANKS
Un composé candidat antagoniste de la protéine ESM-1 peut être sélectionné selon le procédé ci-dessus en tant que produit d'expression d'un insert d'ADN contenu dans un vecteur de phage selon la technique décrite par PARMLEY & SMITH (1988). Dans ce type de banques peptidiques, les inserts d'ADN codent pour des peptides de 8 à 20 acides aminés de longueur, comme cela est décrit par OLDENBURG KR et al. (1992), VALADON P et al. (1996), LUCAS AH (1994), WESTERINK (1995), FELICI et al. (1991 ).A candidate antagonist compound of the ESM-1 protein can be selected according to the above method as an expression product of a DNA insert contained in a phage vector according to the technique described by PARMLEY & SMITH (1988) . In this type of peptide library, the DNA inserts encode peptides of 8 to 20 amino acids in length, as described by OLDENBURG KR et al. (1992), VALADON P et al. (1996), LUCAS AH (1994), WESTERINK (1995), FELICI et al. (1991).
Selon ce mode de réalisation particulier, les phages recombinants exprimant une protéine capable de se fixer sur le polypeptide constitué de la protéine ESM-1 ou un fragment de celle-ci est retenu et le complexe formé entre la protéine ESM-1 ou un fragment de celle-ci et le phage recombinant peut être ultérieurement immunoprécipité par un anticorps monoclonal ou polyclonal anti-ESM-1.According to this particular embodiment, the recombinant phages expressing a protein capable of binding to the polypeptide consisting of the protein ESM-1 or a fragment thereof is retained and the complex formed between the protein ESM-1 or a fragment of this and the recombinant phage can be subsequently immunoprecipitated by a monoclonal or polyclonal antibody anti-ESM-1.
B. COMPOSE CANDIDAT OBTENU PAR DES EXPERIENCES DE COMPETITIONB. CANDIDATE COMPOUND OBTAINED BY COMPETITION EXPERIENCES
Les composés candidats antagonistes de la protéine ESM-1 peuvent être également sélectionnés en ce qu'ils se fixent sur la protéine ESM-1 , ou sur un fragment polypeptidique de celle-ci, en compétition avec un composé antagoniste de la protéine ESM-1 sélectionné antérieurement tels que l'un des anticorps anti-ESM-1 décrits ci-dessus, et tout particulièrement l'anticorps monoclonal sécrété par la lignée d'hybridome MEP08 déposée le 19 Novembre 1997 auprès de la CNCM sous le n° d'accès 1-1941. De telles expériences de compétition sont par exemple décrites dans l'article de BECHARD et al. (2000).The candidate antagonist compounds of the ESM-1 protein can also be selected in that they bind to the ESM-1 protein, or to a polypeptide fragment thereof, in competition with an antagonist compound of the ESM-1 protein previously selected such as one of the anti-ESM-1 antibodies described above, and in particular the monoclonal antibody secreted by the hybridoma line MEP08 deposited on November 19, 1997 with the CNCM under the access number 1-1941. Such competitive experiences are for example described in the article by BECHARD et al. (2000).
C. COMPOSES CANDIDATS ANTAGONISTES DE LA PROTEINE ESM-1 SELECTIONNES PAR CHROMATOGRAPHIE D'AFFINITE.C. ANTAGONIST CANDIDATE COMPOUNDS OF THE ESM-1 PROTEIN SELECTED BY AFFINITY CHROMATOGRAPHY.
Des protéines ou d'autres molécules de toute nature capables de se fixer sur la protéine ESM-1 , ou un fragment polypeptidique de cette protéine, peuvent être sélectionnées en utilisant des colonnes d'affinité sur lesquelles ont été immobilisées la protéine ESM-1 ou un fragment de cette dernière, par exemple en utilisant des techniques classiques, incluant un couplage chimique de la protéine ESM-1 ou d'un fragment de cette dernière à la matrice d'une colonne telle que de l'agarose, ou de l'AffiGel®. Une solution contenant le composé candidat à tester est mis en contact avec le support chromatographique sur lequel est immobilisée la protéine ESM-1 ou un fragment peptidique de celle-ci. Les composés retenus sur la colonne d'affinité sont sélectionnés positivement.Proteins or other molecules of any kind capable of binding to the ESM-1 protein, or a polypeptide fragment of this protein, can be selected using affinity columns on which the ESM-1 protein has been immobilized or a fragment of the latter, for example using conventional techniques, including chemical coupling of the protein ESM-1 or a fragment of the latter to the matrix of a column such as agarose, or Affigel®. A solution containing the candidate compound to be tested is brought into contact with the chromatographic support on which the ESM-1 protein or a peptide fragment thereof is immobilized. The compounds retained on the affinity column are selected positively.
D. COMPOSES CANDIDATS SELECTIONNES PAR DES TECHNIQUES DE BIOCAPTEURS OPTIQUES Un composé candidat antagoniste de la protéine ESM-1 peut également être sélectionné en utilisant un biocapteur optique tel que décrit par EDWARDS et LEATHERBARROW (1997). Cette technique permet la détection d'interactions entre des molécules en temps réel sans nécessiter l'utilisation de molécules marquées. Cette technique est basée sur la résonance de surface du plasmon (SPR pour « Surface Plasmon Résonance »). Brièvement, le composé candidat à tester est fixé sur une surface, telle qu'une matrice de carboxyméthyldextran. Un rayon lumineux est dirigé sur l'extrémité de la surface qui ne contient pas l'échantillon à tester et est réfléchi par cette surface. Le phénomène SPR provoque une réduction d'intensité de la lumière réfléchie avec une association spécifique entre l'angle de la lumière réfléchie et la longueur d'onde du rayon lumineux. La fixation du composé candidat provoque un changement dans l'indice de réfraction de la surface, le changement d'indice de réfraction étant détecté comme une modification du signal SPR.D. CANDIDATE COMPOUNDS SELECTED BY OPTICAL SENSOR TECHNIQUES An ESM-1 protein antagonist candidate compound can also be selected using an optical biosensor as described by EDWARDS and LEATHERBARROW (1997). This technique allows the detection of interactions between molecules in real time without requiring the use of labeled molecules. This technique is based on the surface plasmon resonance (SPR for “Surface Plasmon Resonance”). Briefly, the candidate compound to be tested is attached to a surface, such as a matrix of carboxymethyldextran. A light ray is directed on the end of the surface which does not contain the sample to be tested and is reflected by this surface. The SPR phenomenon causes a reduction in the intensity of the reflected light with a specific association between the angle of the reflected light and the wavelength of the light ray. The binding of the candidate compound causes a change in the refractive index of the surface, the change in refractive index being detected as a modification of the SPR signal.
Un tel procédé de détection par biocapteur optique peut permettre également de sélectionner les composés candidats qui entrent en compétition avec un autre ligand pour la fixation sur la protéine ESM- 1 ou un fragment peptidique de ce dernier.Such an optical biosensor detection method can also make it possible to select the candidate compounds which compete with another ligand for binding to the ESM-1 protein or a peptide fragment of the latter.
Par exemple, un composé candidat antagoniste de la protéine ESM-1 englobe les composés capables d'inhiber la fixation d'un anticorps anti-ESM-1 sur la protéine ESM-1 , d'inhiber la fixation du facteur HGF-SF ou encore des facteurs FGF-2 et FGF-7 sur la protéine ESM-1 ou un fragment peptidique de cette dernière.For example, a candidate antagonist compound of the protein ESM-1 includes the compounds capable of inhibiting the binding of an anti-ESM-1 antibody to the protein ESM-1, of inhibiting the binding of factor HGF-SF or else factors FGF-2 and FGF-7 on the protein ESM-1 or a peptide fragment of the latter.
Ainsi, selon encore un autre aspect, l'invention a trait à un procédé de sélection d'un composé antagoniste de la protéine ESM-1 caractérisé en ce qu'il comprend les étapes suivantes: a) mettre en contact la protéine ESM-1 ou un fragment peptidique de cette dernière en présence de :Thus, according to yet another aspect, the invention relates to a process for the selection of an antagonist compound of the ESM-1 protein, characterized in that it comprises the following steps: a) bringing the ESM-1 protein into contact or a peptide fragment of the latter in the presence of:
(i) un composé antagoniste de la protéine ESM-1 se fixant sur la protéine ESM-1 ; et(i) an ESM-1 protein antagonist compound which binds to the ESM-1 protein; and
(ii) un composé candidat à tester; b) dans une étape séparée de l'étape a), mais éventuellement simultanée à cette dernière, mettre en contact la protéine ESM-1 ou un fragment peptidique de cette dernière avec un composé antagoniste de la protéine ESM-1 se fixant sur la protéine ESM-1 ; c) détecter la quantité respective du composé antagoniste de la protéine ESM-1 fixée à l'issue de chacune des étapes a) et b); et d) sélectionner le composé candidat qui entre en compétition avec le composé antagoniste pour la fixation sur la protéine ESM-1.(ii) a candidate compound to be tested; b) in a step separate from step a), but possibly simultaneous with the latter, bringing the ESM-1 protein or a peptide fragment of the latter with an antagonist compound of the ESM-1 protein binding to the ESM-1 protein; c) detecting the respective amount of the antagonist compound of the ESM-1 protein fixed at the end of each of steps a) and b); and d) selecting the candidate compound which competes with the antagonist compound for binding to the ESM-1 protein.
De préférence, un composé antagoniste de ESM-1 pour la mise en oeuvre du procédé de sélection ci-dessus est un anticorps anti-ESM-1 ou encore un composé antagoniste peptidique tel que défini ci-avant dans la présente description.Preferably, an ESM-1 antagonist compound for carrying out the above selection method is an anti-ESM-1 antibody or else a peptide antagonist compound as defined above in the present description.
Dans un premier mode de réalisation particulier d'un procédé pour sélectionner un composé antagoniste de ESM-1 à partir d'un composé .candidat, ledit procédé comprend les étapes suivantes:In a first particular embodiment of a method for selecting an ESM-1 antagonist compound from a candidate compound, said method comprises the following steps:
1) sélectionner, parmi les composés candidats, les composés qui se fixent sur la protéine ESM-1 ou sur un fragment peptidique de cette protéine;1) select, from the candidate compounds, the compounds which bind to the ESM-1 protein or to a peptide fragment of this protein;
2) administrer un composé sélectionné à l'étape 1 ) à un animal et déterminer la capacité de ce composé à inhiber, chez cet animal, le développement de tumeurs induites par la protéine ESM-1 ; 3) sélectionner les composés inhibiteurs du développement des tumeurs déterminéas à l'étape 2) comme des composés antagonistes de la protéine ESM-1.2) administering a compound selected in step 1) to an animal and determining the capacity of this compound to inhibit, in this animal, the development of tumors induced by the ESM-1 protein; 3) select the compounds inhibiting the development of tumors determined in step 2) as antagonist compounds of the protein ESM-1.
L'étape 1 ) consiste préférentiellement en la mise en oeuvre d'un procédé de sélection d'un composé candidat se fixant sur la protéine ESM-1 ou sur un fragment de celle-ci, choisi parmi les procédés détaillés dans la présente description.Step 1) preferably consists of implementing a process for selecting a candidate compound which binds to the ESM-1 protein or a fragment thereof, chosen from the processes detailed in the present description.
L'étape 2) consiste préférentiellement en la mise en oeuvre d'un procédé de sélection d'un composé candidat in vivo tel que détaillé dans la description. Dans un mode de réalisation particulier du procédé, celui-ci comprend également une étape 4) consistant à sacrifier l'animal. Composition pharmaceutique de l'invention.Step 2) preferably consists of implementing a process for selecting a candidate compound in vivo as detailed in the description. In a particular embodiment of the method, it also comprises a step 4) consisting in sacrificing the animal. Pharmaceutical composition of the invention.
Un autre objet de l'invention consiste en une composition pharmaceutique pour le traitement et/ou la prévention d'un cancer comprenant un composé antagoniste de la protéine ESM-1.Another subject of the invention consists of a pharmaceutical composition for the treatment and / or prevention of cancer comprising a compound antagonist of the protein ESM-1.
Composition pharmaceutique comprenant un composé antagoniste du type anticorps ou du type peptide selon l'invention.Pharmaceutical composition comprising an antagonist compound of the antibody type or of the peptide type according to the invention.
Selon un premier aspect, une composition pharmaceutique selon l'invention comprend une quantité thérapeutiquement efficace d'un anticorps anti-ESM-1 ou d'un composé antagoniste peptidique dérivé de ESM-1 , en association avec un ou plusieurs véhicules pharmaceutiquement compatibles. Les compositions pharmaceutiques selon l'invention incluent celles adaptées pour une administration topique, orale, rectale, nasale ou parentérale (y compris intramusculaire, sous-cutanée et intraveineuse) ou encore sous une forme adaptée pour une administration par inhalation ou insufflation . Les compositions pharmaceutiques selon l'invention peuvent être présentées sous forme d'unités de dosage et peuvent être préparées par toute méthode bien connue de l'homme du métier du domaine de la galénique pharmaceutique. Toutes les méthodes comprennent une étape consistant à associer le composé agoniste constituant le principe actif de la composition avec un véhicule liquide ou un véhicule solide finement divisé et, si nécessaire, mettre en forme le produit, par exemple. sous forme de comprimés ou de gélules.According to a first aspect, a pharmaceutical composition according to the invention comprises a therapeutically effective amount of an anti-ESM-1 antibody or of a peptide antagonist compound derived from ESM-1, in combination with one or more pharmaceutically compatible vehicles. The pharmaceutical compositions according to the invention include those suitable for topical, oral, rectal, nasal or parenteral administration (including intramuscular, subcutaneous and intravenous) or also in a form suitable for administration by inhalation or insufflation. The pharmaceutical compositions according to the invention can be presented in the form of dosage units and can be prepared by any method well known to those skilled in the art in the pharmaceutical pharmaceutical field. All the methods include a step consisting in associating the agonist compound constituting the active principle of the composition with a liquid vehicle or a finely divided solid vehicle and, if necessary, shaping the product, for example. in the form of tablets or capsules.
Pour une administration orale, une composition pharmaceutique selon l'invention est de préférence présentée sous la forme d'unités de dosages telles que des capsules, des comprimés ou des gélules. Lorsqu'elles se présentent sous une forme incluse dans un conteneur pressurisé, la composition pharmaceutique peut comprendre un agent propulseur tel que le dichlorodifluorométhane, le trichlorofluorométhane, le dichlorotétrafluoroéthane, le dioxyde de carbone ou d'autres gaz appropriés. Dans le cas d'un aérosol pressurisé, l'unité de dosage peut être pourvue d'une soupape capable de libérer une quantité donnée de la composition pharmaceutique.For oral administration, a pharmaceutical composition according to the invention is preferably presented in the form of dosage units such as capsules, tablets or capsules. When presented in a form included in a pressurized container, the pharmaceutical composition may include a propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gases. In the case of a pressurized aerosol, the dosing unit can be provided with a valve capable of releasing a given amount of the pharmaceutical composition.
Selon un autre aspect, la composition pharmaceutique selon l'invention peut être sous la forme d'une composition de poudre sèche pour l'administration par inhalation ou insufflation, par exemple sous la forme d'un mélange de poudre du composé antagoniste et d'une poudre base adaptée, telle que le lactose ou l'amidon. La composition de poudre peut être présentée en unité de dosage, par exemple sous la forme de gélules ou de cartouches à partir desquelles la poudre peut être administrée à l'aide d'un dispositif inhalateur ou insufflateur.According to another aspect, the pharmaceutical composition according to the invention may be in the form of a dry powder composition for administration by inhalation or insufflation, for example in the form of a mixture of powder of the antagonist compound and of a suitable base powder, such as lactose or starch. The powder composition can be presented in dosage units, for example in the form of capsules or cartridges from which the powder can be administered using an inhaler or insufflator device.
Un véhicule solide pharmaceutiquement compatible d'une composition pharmaceutique selon l'invention englobe des substances telles que les agents de saveur, les lubrifiants, les solubilisants, les agents de suspension, les agents de charge, les auxiliaires de compression, les liants ou des agents désintégrants ainsi que les matériaux d'encapsulation. Dans les poudres, le véhicule est un solide finement divisé qui est en mélange avec le composé antagoniste de ESM-1 également sous la forme finement divisée. Dans les comprimés, le principe actif antagoniste de ESM-1 est mélangé avec un véhicule ayant des propriétés de compression adaptées et compactées sous la forme et la taille désirées. Les poudres et les comprimés contiennent de préférence moins de 99% du principe actif. Des véhicules solides préférés sont par exemple le phosphate de calcium, le stéarate de magnésium, le talc, les sucres, le lactose, la dextrine, l'amidon, la gélatine, la cellulose, la polyvinylpyrrolidone et les résines d'échange d'ions.A pharmaceutically compatible solid vehicle of a pharmaceutical composition according to the invention includes substances such as flavoring agents, lubricants, solubilizers, suspending agents, bulking agents, compression aids, binders or agents disintegrants as well as encapsulation materials. In powders, the vehicle is a finely divided solid which is mixed with the antagonist compound of ESM-1 also in the finely divided form. In the tablets, the antagonistic active principle of ESM-1 is mixed with a vehicle having suitable compression properties and compacted in the desired shape and size. The powders and tablets preferably contain less than 99% of the active principle. Preferred solid vehicles are for example calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, polyvinylpyrrolidone and ion exchange resins .
Des véhicules liquides sont utilisés pour préparer une composition pharmaceutique selon l'invention sous la forme d'une solution, une suspension, d'une émulsion, d'un sirop, d'un elixir et d'une composition pressurisée. Le principe actif antagoniste de la protéine ESM-1 peut être dissous ou suspendu dans un véhicule pharmaceutiquement acceptable tel que de l'eau, d'un solvant organique, d'un mélange des deux ou des huiles ou matières grasses pharmaceutiquement acceptables. Le véhicule liquide peut contenir d'autres additifs pharmaceutiquement acceptables tels que des agents solubilisants, des émulsifiants , des tampons, des conservateurs, des édulcorants, des agents aromatiques, des agents de suspension, des agents épaississants, des agents colorants, des régulateurs de viscosité, des stabilisants ou des osmo-régulateurs. Des exemples illustratifs de véhicules liquides pour administration orale et parentérale comprennent l'eau, les alcools (y compris les alcools monohydriques et polyhydriques comme les glycols), les huiles telles que l'huile de coco ou l'huile d'arachide fractionnée. Pour l'administration parentérale, le véhicule peut aussi être un ester tel que l'oléate d'éthyle et le myristate d'isopropyle. Les compositions pharmaceutiques liquides sous la forme de solutions ou suspensions stériles peuvent être utilisées pour une injection intramusculaire, intrapéritonéale ou sous-cutanée.Liquid vehicles are used to prepare a pharmaceutical composition according to the invention in the form of a solution, a suspension, an emulsion, a syrup, an elixir and a pressurized composition. The antagonist active principle of the protein ESM-1 can be dissolved or suspended in a pharmaceutically acceptable vehicle such as water, an organic solvent, a mixture of the two or pharmaceutically acceptable oils or fats. The liquid vehicle may contain other pharmaceutically acceptable additives such as agents solubilizers, emulsifiers, buffers, preservatives, sweeteners, aromatics, suspending agents, thickening agents, coloring agents, viscosity regulators, stabilizers or osmo-regulators. Illustrative examples of liquid vehicles for oral and parenteral administration include water, alcohols (including monohydric and polyhydric alcohols such as glycols), oils such as coconut oil or fractionated peanut oil. For parenteral administration, the vehicle can also be an ester such as ethyl oleate and isopropyl myristate. Liquid pharmaceutical compositions in the form of sterile solutions or suspensions can be used for intramuscular, intraperitoneal or subcutaneous injection.
De préférence, une composition pharmaceutique selon l'invention comprend de 1 à 1000 mg de composés antagonistes de la protéine ESM-1 par unité de dosage, et de préférence de 10 à 500 mg du composé antagoniste de la protéine ESM-1 par unité de dosage.Preferably, a pharmaceutical composition according to the invention comprises from 1 to 1000 mg of antagonist compounds of the ESM-1 protein per dosage unit, and preferably from 10 to 500 mg of the antagonist compound of the ESM-1 protein per unit of dosage dosage.
La présente invention concerne aussi une méthode de traitement et/ou de prévention d'un cancer comprenant une étape au cours de laquelle on administre à un patient ayant besoin d'un tel traitement une composition pharmaceutique telle que définie ci-dessus.The present invention also relates to a method of treatment and / or prevention of cancer comprising a step during which a pharmaceutical composition as defined above is administered to a patient in need of such treatment.
Composition pharmaceutique comprenant un composé antagoniste de la protéine ESM-1 du type polynucléotide antisens.Pharmaceutical composition comprising an antagonist compound of the ESM-1 protein of the antisense polynucleotide type.
Font également partie de l'invention des compositions pharmaceutiques contenant une quantité thérapeutiquement efficace d'un composé antagoniste de la protéine ESM-1 du type polynucléotide antisens tel que défini dans la présente description ainsi que des méthodes de traitement et/ou de prévention d'un cancer comprenant l'administration à un patient ayant besoin d'un tel traitement d'une composition pharmaceutique comprenant un polynucléotide antisens tel que défini ci-dessus.Also part of the invention are pharmaceutical compositions containing a therapeutically effective amount of a protein antagonist compound ESM-1 of the antisense polynucleotide type as defined in the present description as well as methods of treatment and / or prevention of a cancer comprising the administration to a patient in need of such treatment of a pharmaceutical composition comprising an antisense polynucleotide as defined above.
Un oligonucléotide antisens selon l'invention peut être administré par tout moyen, qu'il soit local ou systémique. L'administration locale d'un polynucléotide antisens de l'invention, par exemple au niveau d'une tumeur, peut être réalisée par l'administration du polynucléotide antisens directement au niveau de la tumeur ou au niveau du tissu entourant la tumeur et permettant à l'oligonucléotide de migrer et, le cas échéant d'entrer, dans les cellules tumorales. Par exemple, les polynucléotides antisens peuvent être injectés à l'aide d'une seringue. L'injection peut être intramusculaire, intraveineuse, intrapéritonéale ou sous-cutanée. Le polynucléotide antisens peut être administré au foie par l'intermédiaire du système de la veine porte hépatique. De même, le polynucléotide antisens peut être administré au poumon à l'aide d'un dispositif d'inhalation.An antisense oligonucleotide according to the invention can be administered by any means, whether local or systemic. The local administration of an antisense polynucleotide of the invention, for example at the level of a tumor, can be carried out by the administration of the antisense polynucleotide directly at the level of the tumor or at the level of the tissue surrounding the tumor and allowing the oligonucleotide to migrate and, where appropriate, to enter the tumor cells. For example, antisense polynucleotides can be injected using a syringe. The injection can be intramuscular, intravenous, intraperitoneal or subcutaneous. The antisense polynucleotide can be administered to the liver through the hepatic portal vein system. Likewise, the antisense polynucleotide can be administered to the lung using an inhalation device.
D'autres modes d'administration d'un polynucléotide antisens peuvent être utilisés. Par exemple, les polynucléotides antisens peuvent être administrés de manière systémique après leur insertion dans un vecteur d'expression. Le terme « vecteur d'expression », englobe un plasmide, un virus ou un autre véhicule connu dans l'état de la technique pour assurer l'expression d'un polynucléotide antisens.Other modes of administration of an antisense polynucleotide can be used. For example, the antisense polynucleotides can be administered systemically after their insertion into an expression vector. The term "expression vector" encompasses a plasmid, a virus or another vehicle known in the art for ensuring the expression of an antisense polynucleotide.
Pour la mise en oeuvre de vecteurs adaptés pour l'expression recombinante d'un polynucléotide antisens, l'homme du métier pourra avantageusement utiliser des vecteurs pMSXND décrits par LEE et NATHANS (1988), des vecteurs viraux eucaryotes, tels que ceux décrits par GLUZMAN (1982), ou encore des adénovirus et des virus adéno- associés tels que ceux décrits dans les brevets US n°5, 173,414 et 5,354,678 ou encore un système d'expression incluant le vecteur d'expression décrit par MOXHAM et al. (1993).For the implementation of vectors suitable for the recombinant expression of an antisense polynucleotide, those skilled in the art may advantageously use pMSXND vectors described by LEE and NATHANS (1988), eukaryotic viral vectors, such as those described by GLUZMAN (1982), or adenoviruses and adeno-associated viruses such as those described in US patents 5, 173,414 and 5,354,678 or also an expression system including the expression vector described by MOXHAM et al. (1993).
De préférence, le vecteur d'expression contient un promoteur permettant la production du polynucléotide antisens chez un animal, de préférence un mammifère, et de manière préférée chez l'homme, tel que le promoteur de la polyhédrine . Le vecteur d'expression peut être adapté à l'expression ciblée du polynucléotide antisens au site de la tumeur, par exemple en plaçant l'acide nucléique codant pour les polynucléotides antisens sous le contrôle d'un promoteur spécifique de certaines cellules, telles que les cellules épithéliales ou les cellules endothéliales. Un exemple d'un tel promoteur est le promoteur viral désigné NuNTV qui est utile spécifiquement dans le traitement des cancers du sein. D'autres exemples de tels promoteurs spécifiques sont des promoteurs de protéine du lait telle que la β-lactoglobuline, de la caséine-α et de la caséine-β. La quantité thérapeutiquement efficace d'un polynucléotide antisens de l'invention peut être déterminée comme la quantité nécessaire à une réduction significative de la traduction de la protéine ESM-1 au niveau systémique ou local.Preferably, the expression vector contains a promoter allowing the production of the antisense polynucleotide in an animal, preferably a mammal, and more preferably in humans, such as the polyhedrin promoter. The expression vector can be adapted to the targeted expression of the antisense polynucleotide at the tumor site, for example by placing the nucleic acid coding for the antisense polynucleotides under the control of a promoter specific for certain cells, such as epithelial cells or endothelial cells. An example of such a promoter is the designated viral promoter NuNTV which is useful specifically in the treatment of breast cancer. Other examples of such specific promoters are promoters of milk protein such as β-lactoglobulin, casein-α and casein-β. The therapeutically effective amount of an antisense polynucleotide of the invention can be determined as the amount necessary for a significant reduction in the translation of the protein ESM-1 at the systemic or local level.
Il apparaîtra clairement à l'homme du métier que la concentration du polynucléotide antisens thérapeutiquement efficace varie avec le choix de la forme d'administration. Par exemple, si le polynucléotide antisens est administré par injection à un mammifère, l'unité -de dosage constituée d'une seringue contenant une quantité efficace du polynucléotide antisens. Une quantité efficace du polynucléotide antisens pour une administration systémique est comprise entre 0,01 mg/kg à 50 mg/kg administrée une fois ou deux fois par jour. Une quantité thérapeutiquement efficace d'un polynucléotide antisens selon l'invention inclue dans une composition pharmaceutique est comprise généralement entre 104 et 1011 molécules de polynucléotide antisens par administration et de préférence entre 105 et 1010 molécules d'ADN par administration.It will be clear to those skilled in the art that the concentration of the therapeutically effective antisense polynucleotide varies with the choice of the form of administration. For example, if the antisense polynucleotide is administered by injection to a mammal, the dosing unit consisting of a syringe containing an effective amount of the antisense polynucleotide. An effective amount of the antisense polynucleotide for systemic administration is between 0.01 mg / kg to 50 mg / kg administered once or twice a day. A therapeutically effective amount of an antisense polynucleotide according to the invention included in a pharmaceutical composition is generally between 10 4 and 10 11 molecules of antisense polynucleotide per administration and preferably between 10 5 and 10 10 DNA molecules per administration.
Cependant, différents protocoles de dosage peuvent être utilisés selon (i) la capacité individuelle du polynucléotide antisens à inhiber l'expression de la protéine ESM-1 , (ii) la gravité ou l'étendue de la maladie, ou (iii) le comportement pharmacocinétique du polynucléotide antisens utilisé.However, different assay protocols can be used depending on (i) the individual ability of the antisense polynucleotide to inhibit expression of the ESM-1 protein, (ii) the severity or extent of the disease, or (iii) behavior pharmacokinetics of the antisense polynucleotide used.
Le polynucléotide antisens peut être associé à un véhicule pharmaceutiquement compatible ou à un excipient. Des exemples d'excipients comprennent les agents de charge, les liants, les agents désintégrants, les lubrifiants, selon la nature de l'administration et les formes de dosage. Des formes de dosage préférées consistent en des solutions liquides, avantageusement des tampons physiologiquement compatibles telles que des solutions de HANK's ou de RINGER. De plus, les polynucléotides antisens selon l'invention peuvent être formulés sous forme solide puis redissous ou resuspendus immédiatement avant usage. Selon englobe les formes lyophilisées et les liposomes contenant de tels polynucléotides antisens.The antisense polynucleotide can be associated with a pharmaceutically compatible vehicle or with an excipient. Examples of excipients include bulking agents, binders, disintegrants, lubricants, depending on the nature of the administration and the dosage forms. Preferred dosage forms consist of liquid solutions, preferably physiologically compatible buffers such as HANK's or RINGER solutions. In addition, the antisense polynucleotides according to the invention can be formulated in solid form and then redissolved or resuspended immediately before use. According to encompasses lyophilized forms and liposomes containing such antisense polynucleotides.
Un polynucléotide antisens de l'invention peut aussi être administré de manière systémique par les voies transmuqueuses, transdermiques ou orales. Pour les voies d'administration transmuqueuses ou transdermiques, des agents pénétrant peuvent être utilisés dans la formulation tels que des sels biliaires ou des dérivés de l'acide fusidique.An antisense polynucleotide of the invention can also be administered systemically by the transmucosal, transdermal or oral routes. For transmucosal or transdermal routes of administration, penetrating agents can be used in the formulation such as bile salts or fusidic acid derivatives.
La présente invention est également relative à une méthode de traitement et/ou de prévention d'un cancer comprenant une étape d'administration à un patient, ayant besoin d'un tel traitement, une composition pharmaceutique telle que définie ci-dessus comprenant un composé antagoniste de ESM-1 du type polynucléotide antisens.. .. .The present invention also relates to a method of treatment and / or prevention of cancer comprising a step of administration to a patient, in need of such treatment, a pharmaceutical composition as defined above comprising a compound ESM-1 antagonist of the antisense polynucleotide type .. ...
De manière générale, l'une quelconque des compositions pharmaceutiques de l'invention telles que définies ci-dessus et comprenant une quantité thérapeutiquement efficace d'un composé antagoniste de la protéine ESM-1 est utile dans la prévention et/ou le traitement d'un cancer.Generally, any of the pharmaceutical compositions of the invention as defined above and comprising a therapeutically effective amount of an ESM-1 protein antagonist compound is useful in the prevention and / or treatment of cancer.
A titre illustratif, mais non limitatif, une composition pharmaceutique selon l'invention est utile pour la prévention et/ou le traitement de cancers tels que les cancers des voies respiratoires, les cancers broncho-pulmonaires, les cancers du sein, les cancers du colon et les cancers rénaux ainsi que les cancers des voies digestives.By way of illustration, but not limitation, a pharmaceutical composition according to the invention is useful for the prevention and / or treatment of cancers such as cancers of the respiratory tract, bronchopulmonary cancers, breast cancers, colon cancers and renal cancers as well as cancers of the digestive tract.
La présente invention est en outre illustrée, sans pour autant être limitée, par les figures et les exemples suivants.The present invention is further illustrated, without being limited, by the following figures and examples.
FIGURESFIGURES
La figure 1 illustre des gels d'immuno-empreinte (« Western- Blot ») et des colorations de ESM-1 sur gel SDS-PAGE. Chaque gel d'immuno-empreinte a été révélé avec l'anticorps monoclonal anti-ESM-1 MEP14. Le second anticorps anti-souris marqué à la peroxydase de raifort a été purifié par affinité et donne des résultats négatifs lorsqu'il est utilisé seul. Figure 1A. Gel d'immuno-empreinte de la protéine ESM-1 de différents types cellulaires exprimant cette protéine.FIG. 1 illustrates immunoblotting gels (“Western-Blot”) and stains of ESM-1 on SDS-PAGE gel. Each immunoblock gel was revealed with the anti-ESM-1 MEP14 monoclonal antibody. The second anti-mouse antibody labeled with horseradish peroxidase has been affinity purified and gives negative results when used alone. Figure 1A. ESM-1 protein immunoblot gel of different cell types expressing this protein.
L'immuno-précipitation de ESM-1 à partir des surnageants de culture des cellules SVI (1), 293-ESM(2) et CHO-ESM(3) a été réalisée avec l'anticorps MEP19 lorsque cela est indiqué, ou avec un anticorps témoin. Les flèches indiquent la bande spécifique de ESM-1. La forme native de ESM-1 est représentée par une bande diffuse aux alentours de 50 kD.Immunoprecipitation of ESM-1 from culture supernatants of cells SVI (1), 293-ESM (2) and CHO-ESM (3) was carried out with the antibody MEP19 when indicated, or with a control antibody. The arrows indicate the specific band of ESM-1. The native form of ESM-1 is represented by a diffuse band around 50 kD.
Figure 1B. Absence de détection de la protéine purifiée ESM-1 avec le bleu de coomassie.Figure 1B. Absence of detection of the purified protein ESM-1 with coomassie blue.
5 μg de protéine ESM-1 purifiée à partir des cellules SVI ont été chargés sur un gel SDS-PAGE à 15% et colorés au bleu de coomassie afin de détecter le corps peptidique de la molécule. Les flèches indiquent l'absence de détection de ESM-1.5 μg of protein ESM-1 purified from SVI cells were loaded onto a 15% SDS-PAGE gel and stained with coomassie blue in order to detect the peptide body of the molecule. The arrows indicate the absence of ESM-1 detection.
Figure 1C. Détection de la protéine ESM-1 purifiée avec le bleu alcian.Figure 1C. Detection of the protein ESM-1 purified with alkian blue.
5 μg de protéine ESM-1 purifiée à partir des cellules SVI ont été chargés sur un gel SDS-PAGE à 15% et ont été révélés par coloration au bleu alcian afin de détecter le corps glycanique de la molécule. La flèche indique la protéine ESM-1.5 μg of protein ESM-1 purified from SVI cells were loaded onto a 15% SDS-PAGE gel and were revealed by alcian blue staining in order to detect the glycan body of the molecule. The arrow indicates the protein ESM-1.
La figure 2 illustre le poids moléculaire apparent des corps peptidique et glycanique de ESM-1.FIG. 2 illustrates the apparent molecular weight of the peptide and glycan bodies of ESM-1.
Figure 2A. Analyse par mutation du site de fixation de O- glycosylation.Figure 2A. Analysis by mutation of the O-glycosylation binding site.
Deux sites putatifs de O-glycosylation (tréonine 120 et serineTwo putative O-glycosylation sites (treonin 120 and serine
137) ont été substitués par un résidu alanine par mutagénèse dirigée. La protéine ESM-1 sauvage (WT pour « Wild-Type »), les mutants de ESM- 1 T120A et S137A) ainsi que les témoins négatifs (MOCK) ont été transfectés dans les cellules 293 et les surnageants de culture cellulaire ainsi que les lysats cellulaires ont été analysés par immuno-empreinte (Western-Blot) à l'aide de l'anticorps monoclonal MEP14. Les flèches indiquent les bandes spécifiques.137) were substituted with an alanine residue by site-directed mutagenesis. The wild ESM-1 protein (WT for “Wild-Type”), the ESM-1 mutants T120A and S137A) as well as the negative controls (MOCK) were transfected into 293 cells and the cell culture supernatants as well as the cell lysates were analyzed by immunoblotting (Western-Blot) using the monoclonal antibody MEP14. The arrows indicate the specific bands.
Figure 2B. Effet d'un traitement à la proteinase K sur ESM-Figure 2B. Effect of proteinase K treatment on ESM-
1.1.
La protéine ESM-1 purifiée à partir des cellules SVI(1 ) et 293- ESM (2) a été digérée par la proteinase K et chargée sur un gel SDS- PAGE 15%. La flèche supérieure indique la forme sauvage de la protéine ESM-1 non traitée et la flèche inférieure indique la protéine ESM-1 digérée par la proteinase K.The ESM-1 protein purified from SVI (1) and 293-ESM (2) cells was digested with proteinase K and loaded onto a 15% SDS-PAGE gel. The upper arrow indicates the wild form of the untreated ESM-1 protein and the lower arrow indicates the proteinase K digested ESM-1 protein.
La figure 3 illustre les effets de chondroïtinases spécifiques sur ESM-1.Figure 3 illustrates the effects of specific chondroitinases on ESM-1.
Figure 3A. Traitement de la protéine ESM-1 sauvage purifiée avec la chondroïtinase ABC.Figure 3A. Treatment of purified wild protein ESM-1 with chondroitinase ABC.
La protéine ESM-1 sécrétée a été purifiée par chromatographie d'échanges d'ions, suivie d'une chromatographie d'immuno-affinité à partir de surnageants de cultures de cellules SVI(1 ), 293-ESM(2) et à partir de plasma humain (3), puis digérée ou non par la chondroïtinase ABC. 50 ng de la protéine digérée ont été chargés sur un gel SDS-PAGE à 15% puis analysés par immuno-empreinte (Western-Blot). La flèche supérieure indique les formes non digérées de ESM-1 et la flèche inférieure indique les formes digérées de ESM-1.The secreted ESM-1 protein was purified by ion exchange chromatography, followed by immunoaffinity chromatography from supernatants from SVI (1), 293-ESM (2) cell cultures and from of human plasma (3), then digested or not by chondroitinase ABC. 50 ng of the digested protein were loaded onto a 15% SDS-PAGE gel and then analyzed by immunoblotting (Western-Blot). The upper arrow indicates undigested forms of ESM-1 and the lower arrow indicates digested forms of ESM-1.
Figure 3B. Traitement de la protéine ESM-1 sauvage purifiée avec la chondroïtinase B.Figure 3B. Treatment of purified wild-type ESM-1 protein with chondroitinase B.
La protéine ESM-1 purifiée à partir de surnageants de culture des cellules SVI (1 ) et 293-ESM(2) ont été digérés ou non par la chondroïtinase. Les protéines ont été chargées sur un gel SDS-PAGE à 15% . La flèche supérieure indique les différentes formes de ESM-1 non digérées autour de 50 kD et la flèche inférieure indique les différentes formes de la protéine ESM-1 digérée, autour de 22 kD.The ESM-1 protein purified from culture supernatants of SVI (1) and 293-ESM (2) cells were digested or not by chondroitinase. Proteins were loaded onto a 15% SDS-PAGE gel. The upper arrow indicates the different forms of ESM-1 no digested around 50 kD and the lower arrow indicates the different forms of the digested ESM-1 protein, around 22 kD.
Figure 3C. Traitement de la protéine ESM-1 sauvage purifiée avec la chondroïtinase AC.Figure 3C. Treatment of purified wild protein ESM-1 with chondroitinase AC.
La protéine ESM-1 purifiée à partir de surnageants de culture des cellules HUVEC (1) et 293-ESM(2) ont été virées??? par la chondroïtinase AC et ont été chargées sur un gel SDS-PAGE à 15%. La flèche supérieure indique les différentes formes de ESM-1 non digérées autour de 502 kD et la flèche inférieure indique les différentes formes digérées de la protéine ESM-1 autour de 22 kD.The protein ESM-1 purified from culture supernatants of cells HUVEC (1) and 293-ESM (2) have been fired ??? with chondroitinase AC and were loaded onto a 15% SDS-PAGE gel. The upper arrow indicates the different forms of undigested ESM-1 around 502 kD and the lower arrow indicates the different digested forms of the ESM-1 protein around 22 kD.
Figure 3D. Traitement de la protéine ESM-1 sauvage. purifiée avec la chondroïtinase C.Figure 3D. Treatment of wild ESM-1 protein. purified with chondroitinase C.
La protéine ESM-1 purifiée à partir de surnageants de cultures des cellules HUVEC (1) et 293-ESM(2) ont été digérées ou non par la chondroïtinase C et ont été chargées sur un gel SDS-PAGE à 15%. La flèche supérieure indique les différentes formes non digérées de ESM-14 autour de 50 kD et la flèche inférieure indique les différentes formes digérées de la protéine ESM-1 autour de 22 kD.The protein ESM-1 purified from culture supernatants from HUVEC (1) and 293-ESM (2) cells were digested or not by chondroitinase C and were loaded onto a 15% SDS-PAGE gel. The upper arrow indicates the different undigested forms of ESM-14 around 50 kD and the lower arrow indicates the different digested forms of the ESM-1 protein around 22 kD.
La figure 4 illustre les effets de la protéine ESM-1 sauvage purifiée sur le temps de coagulation en présence de thrombine. On peut noter le retard et la réduction dans la formation de thrombine pour le plasma héparine ainsi que pour les quatre autres courbes (plasma riche en plaquette ou PRP: losange vide; plasma riche en plaquette + ESM-1 à 0,2 mg/ml: carré plein; plasma riche en plaquette + ESM-1 0 0,5 mg/ml: triangle plein; plasma riche en plaquette + ESM-1 à 1 mg/ml : cercle plein; plasma riche en plaquette + héparine: cercle vide).FIG. 4 illustrates the effects of the purified wild-type ESM-1 protein on the coagulation time in the presence of thrombin. The delay and reduction in thrombin formation can be noted for the heparin plasma as well as for the other four curves (plasma rich in platelet or PRP: empty diamond; plasma rich in platelet + ESM-1 at 0.2 mg / ml : full square; plasma rich in platelet + ESM-1 0 0.5 mg / ml: solid triangle; plasma rich in platelet + ESM-1 at 1 mg / ml: full circle; plasma rich in platelet + heparin: empty circle) .
La figure 5 illustre l'activité biologique du protéoglycane ESM-1 sur la prolifération des cellules 293 induites par le facteur HGF/SF. La stimulation de l'incorporation de 3H-thymidine par les cellules 293 induites par le facteur HGF/SF a été étudiée. Les cellules ont été ensemencées à 1 x 104 cellules par puits dans du milieu DMEM additionnées de transférine et d'insuline et de HGF/SF à 50 ng par ml avant addition de différentes molécules. Les barres représentent le pourcentage d'augmentation de l'incorporation de 3H-thymidine (moyenne +/- S.E.M. d'échantillons en triple d'une expérience représentative) en présence des additions indiquées de sérum, des différentes formes de ESM-1 à 2,5 mg/ml et de décorine à 2,5 mg/ml. Le bruit de fond d'incorporation de 3H-thymidine en présence de HGF/SF a été généralement compris entre 7.000 et 8.000 cpm par puits. Les résultats présentés sont similaires à ceux obtenus dans trois autres expériences séparées.FIG. 5 illustrates the biological activity of the proteoglycan ESM-1 on the proliferation of 293 cells induced by the HGF / SF factor. The stimulation of the incorporation of 3 H-thymidine by the 293 cells induced by the HGF / SF factor was studied. The cells were seeded at 1 × 10 4 cells per well in DMEM medium added transferin and insulin and HGF / SF at 50 ng per ml before adding different molecules. The bars represent the percentage increase in the incorporation of 3 H-thymidine (mean +/- SEM of samples in triplicate from a representative experiment) in the presence of the indicated additions of serum, of the different forms of ESM-1 to 2.5 mg / ml and decorin at 2.5 mg / ml. The background noise of incorporation of 3 H-thymidine in the presence of HGF / SF was generally between 7,000 and 8,000 cpm per well. The results presented are similar to those obtained in three other separate experiments.
La figure 6 illustre une étude de douze réponses des différentes formes de ESM-1 et de décorine sur l'activité mitogène induite par le facteur HGF/SF. On a étudié la simulation de la synthèse d'ADN par les cellules 293 en présence de HGF/SF à 50 nanogrammes par ml seule ou en présence de différentes concentrations de la protéine sauvage ESM- 1/WT (carré vide), de la protéine mutée non glycosylée ESM/S137A (cercle plein), de la chaîne GAG dérivée de la protéine ESM/WT (carré plein) ou de la décorine (cercle vide). Les valeurs moyennes de mesure en triple d'incorporation de 3H-thymidine obtenues dans une expérience parmi trois expériences indépendantes sont représentées à la figure 6. Les résultats sont exprimés en cpm. Les déviations standard sont approximativement de 10%.FIG. 6 illustrates a study of twelve responses of the different forms of ESM-1 and decorin on the mitogenic activity induced by the HGF / SF factor. We studied the simulation of DNA synthesis by 293 cells in the presence of HGF / SF at 50 nanograms per ml alone or in the presence of different concentrations of the wild protein ESM-1 / WT (empty square), of the protein non-glycosylated mutant ESM / S137A (solid circle), of the GAG chain derived from the protein ESM / WT (solid square) or of decorin (empty circle). The mean measurement values in triplicate of incorporation of 3 H-thymidine obtained in an experiment from three independent experiments are shown in FIG. 6. The results are expressed in cpm. Standard deviations are approximately 10%.
La figure 7 illustre le pouvoir tumorigène de la protéine ESM-1. Deux lots de plus de 10 souris ont reçu des cellules HEK témoins ou des cellules HEK transfectees avec un vecteur codant pour l'ADNc de la protéine ESM-1 sauvage (ESM/WT). Sur la figure 7A, le pourcentage de tumeurs macroscopiquement visibles à la huitième semaine au point d'injection et dont le volume tumoral est supérieur à 1 cm3 est représenté en ordonnée . La figure 7B illustre la cinétique d'apparition des tumeurs chez des souris ayant reçu les cellules HEK transfectees exprimant la protéine ESM-1 glycosylée (ESM/WT). Le nombre de semaines après l'injection des cellules est représenté en abscisse. En ordonnée est représenté le volume tumoral moyen, exprimé en cm3.Figure 7 illustrates the tumorigenicity of the protein ESM-1. Two batches of more than 10 mice received HEK control cells or HEK cells transfected with a vector coding for the cDNA of the wild-type ESM-1 protein (ESM / WT). In FIG. 7A, the percentage of macroscopically visible tumors at the eighth week at the injection site and whose tumor volume is greater than 1 cm 3 is represented on the ordinate. FIG. 7B illustrates the kinetics of appearance of tumors in mice having received transfected HEK cells expressing the glycosylated protein ESM-1 (ESM / WT). The number of weeks after the injection of the cells is shown on the abscissa. The ordinate shows the average tumor volume, expressed in cm 3 .
La figure 8 illustre la production de ESM-1 par les tumeurs induites chez la souris. La figure 8A représente le taux sérique de protéine ESM-1 retrouvé chez les deux lots de souris, à la huitième semaine suivant l'injection des cellules. En abscisses sont représentés respectivement le lot de souris ayant reçu les cellules HEK témoins et le lot de souris ayant reçu les cellules HEK exprimant la protéine ESM-1 glycosylée (ESM/WT). Le taux sérique de ESM-1 retrouvé, exprimé en nanogramme/ml, est représenté en ordonnées.Figure 8 illustrates the production of ESM-1 by tumors induced in mice. FIG. 8A represents the serum level of protein ESM-1 found in the two batches of mice, at the eighth week following the injection of the cells. On the abscissa are shown respectively the batch of mice having received the control HEK cells and the batch of mice having received the HEK cells expressing the glycosylated protein ESM-1 (ESM / WT). The serum ESM-1 level found, expressed in nanograms / ml, is represented on the ordinate.
La figure 8B illustre la cinétique des taux sériques de ESM-1 mesurées par ELISA, pour les souris du lot ayant reçu les cellules HEK transfectees avec un ADN codant pour la protéine ESM-1 glycosylée (ESM/WT). En abscisses sont représentés le nombre de semaines suivant l'injection des cellules transfectees. Le taux sérique de protéine ESM-1 , et exprimé en nanogramme/ml, est représenté en ordonnées.FIG. 8B illustrates the kinetics of the serum levels of ESM-1 measured by ELISA, for the mice of the batch having received HEK cells transfected with a DNA coding for the glycosylated protein ESM-1 (ESM / WT). The abscissa shows the number of weeks following the injection of the transfected cells. The serum ESM-1 protein level, expressed in nanograms / ml, is represented on the ordinate.
La figure 9 illustre l'activité tumorigène de différentes formes de la protéine ESM-1.FIG. 9 illustrates the tumorigenic activity of different forms of the protein ESM-1.
La figure 9A illustre l'apparition de tumeurs chez différents lots de souris, les souris ayant reçu respectivement les cellules HEK témoins, les cellules HEK transfectees avec un ADNc codant pour la protéine ESM-1 glycosylée (ESM/WT), les cellules transfectees avec la protéine ESM-1 non glycosylées (ESM/S137A) et les cellules HEK transfectees avec un ADNc codant pour la protéine ESM-1 mutée en positions 134 et 135 (ESM/F1 15A, F116A). En ordonnées est représenté le pourcentage de tumeurs macroscopiquement visibles à la huitième semaine au point d'injection dont le volume tumoral est supérieur à 1 cm3. La figure 9B illustre le taux sérique de ESM-1 chez les différents lots identiques de souris. Le taux sérique de ESM-1 , exprimé en nanogramme/ml, est représenté en ordonnées.FIG. 9A illustrates the appearance of tumors in different batches of mice, the mice having respectively received the HEK control cells, the HEK cells transfected with a cDNA coding for the glycosylated protein ESM-1 (ESM / WT), the cells transfected with the non-glycosylated ESM-1 protein (ESM / S137A) and the HEK cells transfected with a cDNA coding for the ESM-1 protein mutated at positions 134 and 135 (ESM / F1 15A, F116A). The ordinate shows the percentage of macroscopically visible tumors at the eighth week at the injection site, the tumor volume of which is greater than 1 cm 3 . FIG. 9B illustrates the serum level of ESM-1 in the different identical batches of mice. The serum ESM-1 level, expressed in nanograms / ml, is represented on the ordinate.
La figure 10 illustre l'effet inhibiteur de l'anticorps monoclonal MEP08 sur l'activité pro-tumorale de la protéine ESM-1. L'injection d'anticorps MEP-08 augmente la survie des souris du groupe HEK ESM/WT. Les anticorps monoclonaux MEP-08 sont injectés par voie intrapéritonéale à la dose de 400 μg à partir de la deuxième suivant l'inoculation des cellules HEK/ESM-WT. Les injections sont répétées de façon hebdomadaire pendant 12 semaines. Un anticorps contrôle, MEP-14, est utilisé dans les mêmes conditions. Les souris sont sacrifiées lorsque leur volume tumoral est supérieur à 6 cm3, (n > 8 souris dans chaque groupe). La figure représente le pourcentage de souris vivantes dans chacun des groupes.FIG. 10 illustrates the inhibitory effect of the monoclonal antibody MEP08 on the pro-tumor activity of the protein ESM-1. The injection of MEP-08 antibodies increases the survival of the mice of the HEK ESM / WT group. The MEP-08 monoclonal antibodies are injected intraperitoneally at a dose of 400 μg from the second following inoculation of the HEK / ESM-WT cells. The injections are repeated weekly for 12 weeks. A control antibody, MEP-14, is used under the same conditions. The mice are sacrificed when their tumor volume is greater than 6 cm 3 , (n> 8 mice in each group). The figure represents the percentage of live mice in each of the groups.
EXEMPLES: EXEMPLE 1EXAMPLES: EXAMPLE 1
Modification post-traductionnelle de la forme sécrétée de la protéine ESM-1.Post-translational modification of the secreted form of the ESM-1 protein.
A. Matériels et MéthodesA. Materials and Methods
A.1 Culture cellulaire et matérielsA.1 Cell culture and materials
Les cellules CHO ont été cultivées dans un milieu de culture MAMα (Gibco BRL, Life Technologies, France) additionné de 10% de sérum de veau foetal. Les cellules endothéliales humaines transfectees par le virus SV40, les cellules SV1 décrites par LASSALLE P et al. (1992) ont été cultivées dans du milieu RPMI 1640 contenant 2mM de L- glutamine et 10% de sérum de veau foetal. Les cellules de reins embryonnaire humaine, les cellules de la lignée 293, ont été cultivées dans du milieu DMEM de Dulbecco avec 10% de sérum de veau foetal. Les cellules rénales embryonnaires humaines, cellules de la lignée 293, utilisées pour le test de prolifération ont été cultivées dans du milieu de EAGLE modifié de Dulbecco (Gibco BRL) complémenté avec de l'insuline à 10 mg/ml et de la transférine à 10mg/ml. La proteinase et la chondroïtinase ABC sont commercialisée par Boehringer Mannheim. Les chondroïtinases B, AC et C sont commercialisées par Sigma Le facteur HGF/SF humaine est commercialisé par R & D et la décorine est commercialisée par Sigma. Des anticorps monoclonaux anti-ESM-1 ont été produits et purifiés comme décrit par BECHARD et al. (2000).CHO cells were cultured in a MAMα culture medium (Gibco BRL, Life Technologies, France) supplemented with 10% fetal calf serum. Human endothelial cells transfected with the SV40 virus, the SV1 cells described by LASSALLE P et al. (1992) were cultured in RPMI 1640 medium containing 2 mM L-glutamine and 10% fetal calf serum. Human embryonic kidney cells, cells of line 293, were cultured in DMEM medium from Dulbecco with 10% fetal calf serum. The human embryonic renal cells, cells of line 293, used for the proliferation test were cultured in medium of EAGLE modified by Dulbecco (Gibco BRL) supplemented with insulin at 10 mg / ml and with transferin at 10 mg / ml. Proteinase and chondroitinase ABC are marketed by Boehringer Mannheim. Chondroitinases B, AC and C are marketed by Sigma Human HGF / SF factor is marketed by R & D and decorin is marketed by Sigma. Anti-ESM-1 monoclonal antibodies were produced and purified as described by BECHARD et al. (2000).
A.2 Développement de lignées cellulaires exprimant ESM-1.A.2 Development of cell lines expressing ESM-1.
L'ADNc complet codant pour ESM-1 a été dirigé, purifié et inséré dans le vecteur d'expression pcDNA3 (commercialisé par Invitrogen) entre les sites Xhol et Hindllll. La constructions de vecteurs a été transfectée dans des lignées cellulaires CHO et 293 en présence de lipofectamine (GIBCO BRL), puis sélectionnées sur G418 (1000 μg/ml pour la lignée CHO et 300 μg/ml pour la lignée 293). Les lignées cellulaires transfecfées de manière stable ont été obtenues par dilution limite et les cellules ainsi sélectionnées ont été désignées respectivement CHO-ESM et 2936-ESMThe complete cDNA encoding ESM-1 was directed, purified and inserted into the expression vector pcDNA3 (marketed by Invitrogen) between the Xhol and Hindllll sites. The vector constructs were transfected into CHO and 293 cell lines in the presence of lipofectamine (GIBCO BRL), then selected on G418 (1000 μg / ml for the CHO line and 300 μg / ml for the line 293). Stably transfected cell lines were obtained by limiting dilution and the cells thus selected were designated CHO-ESM and 2936-ESM respectively
A.3 Détermination du site de o-glycosylation de ESM-1 par analyse par mutation.A.3 Determination of the o-glycosylation site of ESM-1 by mutation analysis.
2 sites potentiels de O-glycosylation ont été prédits à l'aide du logiciel NET O glyc:0 Prédiction Serveur.2 potential O-glycosylation sites were predicted using NET O glyc software: 0 Server Prediction.
On a substitué le résidu serine en position 137 (SEQ ID N°1 ) et la thréonine en position 120 (SEQ ID N°1) par un résidu alanine. Les mutants de O-glycosylation ont été produits par PCR à l'aide du kit QuickThe serine residue at position 137 (SEQ ID No. 1) and the threonine at position 120 (SEQ ID No. 1) were substituted with an alanine residue. O-glycosylation mutants were produced by PCR using the Quick kit
Change de mutagénèse dirigé selon les recommandations du fabriquantChange of directed mutagenesis according to the manufacturer's recommendations
(Stratagene).(Stratagene).
Les ADNc mutés ont été confirmés par séquençage (sequenceur ABI prism 377 de la Société Applied Biosystems. Puis les cellules 293 ont été transfectees avec les vecteurs dans lesquels ont été insérés les ADNc mutants pour obtenir des transfectants transitoires et des transfectants stables, respectivement les lignées 293-ESM/S 137A et 293-ESM/T120A.The mutated cDNAs were confirmed by sequencing (ABI prism 377 sequencer from the company Applied Biosystems. Then the 293 cells were transfected with the vectors into which the mutant cDNAs were inserted to obtain transient transfectants and stable transfectants, respectively the lines. 293-ESM / S 137A and 293-ESM / T120A.
A.4. Purification du protéoglycane ESM-1 chondroïtine/dermatane sulfate.A.4. Purification of the ESM-1 proteoglycan chondroitin / dermatan sulfate.
Les surnageants de culture cellulaire ont été ajustés à pH8, puis passés sur une colonne de DEAE-Sepharose (Pharmacia), lavés avec un tampon 50 mM Tris (pH8), 0,2 MNaCI, puis élues dans un tampon 50 mM Tris (pH8), 0,8 M Nacl.The cell culture supernatants were adjusted to pH8, then passed over a DEAE-Sepharose column (Pharmacia), washed with a 50 mM Tris buffer (pH8), 0.2 MNaCI, then eluted in a 50 mM Tris buffer (pH8 ), 0.8 M Nacl.
Les éluats ont été ajustés à 50 mM Tris (pH8), 0,5 MNaCI et passés sur une colonne d'affinité. La colonne d'affinité est constituée d'anticorps monoclonaux anti-ESM-1 (produits par la lignée d'hybridome MEC4) immobilisés sur un gel Affigel Hz hydrazide , selon les recommandations du fabricant (Biorad).The eluates were adjusted to 50 mM Tris (pH8), 0.5 MNaCI and passed through an affinity column. The affinity column consists of anti-ESM-1 monoclonal antibodies (produced by the hybridoma line MEC4) immobilized on an Affigel Hz hydrazide gel, according to the manufacturer's recommendations (Biorad).
Après une étape de lavage avec un tampon 50 mM Tris (pH8), MNaCI 0,5, la protéine ESM-1 a été éluée avec une solution de 3M de MgCI2, concentrée et dialysée contre le même tampon sur un dispositif ultrafree 30 (millipore).After a washing step with a 50 mM Tris buffer (pH8), MNaCI 0.5, the ESM-1 protein was eluted with a 3M solution of MgCI 2 , concentrated and dialyzed against the same buffer on an ultrafree device 30 ( millipore).
Le matériau élue a ensuite été quantifié par immunodétection avec des anticorps anti-ESM-1 , et contrôlés sur SDS-PAGE à l'aide d'une coloration au bleu de coomassie ou bleu alcian. La purification de la protéine ESM-1 à partir du plasma humain a été réalisée selon le protocole suivant.The eluted material was then quantified by immunodetection with anti-ESM-1 antibodies, and checked on SDS-PAGE using a staining with coomassie blue or alkian blue. The purification of the ESM-1 protein from human plasma was carried out according to the following protocol.
800 ml de plasma fourni par l'établissement de transfusion sanguine (Lille, France) ont été précipités avec une solution à 60% de sulfate d'ammonium et dialyses contre un tampon 50 mM Tris (pH 8), 0,5 MNAcl. L'extrait de plasma précipité et dialyse a ensuite été passé sur une pré-colonne de 50 ml du type Affigel (Biorad) avant, un passage sur la colonne d'immunoaffinité anti-ESM-1. La protéine ESM-1 fixée sur la colonne d'immunoaffinité a été récupérée comme décrit ci-dessus.800 ml of plasma supplied by the blood transfusion establishment (Lille, France) were precipitated with a 60% solution of ammonium sulfate and dialyzed against a 50 mM Tris buffer (pH 8), 0.5 MNAcl. The precipitated plasma extract and dialysis was then passed over a 50 ml pre-column of the Affigel type (Biorad) before passing through the anti-ESM-1 immunoaffinity column. The ESM-1 protein fixed on the immunoaffinity column was recovered as described above.
La forme non glycosylée de ESM-1 (ESM/S137A) a été purifiée en une seule étape par chromatographie et d'immunoaffinité. Le degré de pureté de la protéine ESM-1 glycosylée (ESM/WT) et de la protéine non glycosylée mutée sur la serine 137 (ESM/S137A) a été contrôlée par FPLC. Le matériau purifié est exempt d'endotoxines, comme en témoigne les résultats du test de lysat de limulus amebocyte (BlOwhitaker).The non-glycosylated form of ESM-1 (ESM / S137A) was purified in a single step by chromatography and immunoaffinity. The degree of purity of the glycosylated protein ESM-1 (ESM / WT) and of the serine 137 non-glycosylated protein (ESM / S137A) was checked by FPLC. The purified material is free of endotoxins, as evidenced by the results of the limulus amebocyte lysate test (BlOwhitaker).
A.5 Immunoprecipitation. immuno-empreinte et séquençage.A.5 Immunoprecipitation. immunoblotting and sequencing.
La taille des différentes formes de ESM-1 a été déterminée par immunoprecipitation et immuno-emprente à partir des surnageants de cultures cellulaires et des lysats cellulaires. Les cellules ont été lysées dans un tampon contenant 0,5% de NP40, un cocktail d'anti-protéases (Boehringer Mannheim, Allemagne) dans du PBS pendant 30 minutes à 4°C en agitation. Puis, les lysats ont été centrifugés à 10.000g pendant 15 min afin d'obtenir des lysats cellulaires clarifiés.The size of the different forms of ESM-1 was determined by immunoprecipitation and immuno-imprinting from cell culture supernatants and cell lysates. The cells were lysed in a buffer containing 0.5% of NP40, a cocktail of anti-proteases (Boehringer Mannheim, Germany) in PBS for 30 minutes at 4 ° C with shaking. Then, the lysates were centrifuged at 10,000g for 15 min to obtain clarified cell lysates.
Les surnageants de culture ont été filtrés sur un filtre ayant un diamètre de pore de 0,45 mm. 1 μg de l'anticorps monoclonal ESM-1 produit par la lignée d'hybridome MEP19 ou 1 μg d'anticorps monoclonal anti-ICAM-1 (clone 164B) a été ajouté au lysat clarifié ou au surnageant de culture cellulaire et incubé pendant une nuit à 4°C sous agitation.The culture supernatants were filtered through a filter having a pore diameter of 0.45 mm. 1 μg of the ESM-1 monoclonal antibody produced by the MEP19 hybridoma line or 1 μg of anti-ICAM-1 monoclonal antibody (clone 164B) was added to the clarified lysate or to the cell culture supernatant and incubated for one overnight at 4 ° C with stirring.
50 μl d'une immunoglobuline anti-souris conjugué à des billes d'agarose (sigma) ont été ajoutés à 4°C pendant 90 min, avant centrifugation et de lavage avec un tampon de lyse et de lavage dans du PBS.50 μl of an anti-mouse immunoglobulin conjugated to agarose beads (sigma) were added at 4 ° C. for 90 min, before centrifugation and washing with a lysis and washing buffer in PBS.
Les billes ont été resuspendues dans 20 et 40 μl de tampon SDS-PAGE pendant 5 min, centrifugées, et les surnageants ont été analysés.The beads were resuspended in 20 and 40 μl of SDS-PAGE buffer for 5 min, centrifuged, and the supernatants were analyzed.
Les échantillons ont été soumis à une électrophorèse sur gel SDS-PAGE, puis transférés sur une membrane de nitrocellulose selon des procédures standard.The samples were subjected to SDS-PAGE gel electrophoresis, then transferred to a nitrocellulose membrane according to standard procedures.
Après une étape de blocage, les membranes ont été incubées pendant une heure avec un anticorps monoclonal ESM-1 produit par la lignée d'hybridome MEP14 à 1 μl, lavées puis incubées pendant I heure avec un anticorps secondaire anti-Fc de souris conjugué à la peroxydase de raifort (« Horse Radish peroxydase ») (commercialisé par la Société SIGMA). On a réalisé plusieurs lavages avant révélation en utilisant le kit de détection ECL commercialisé par la Société Amersham.After a blocking step, the membranes were incubated for one hour with a monoclonal antibody ESM-1 produced by the hybridoma line MEP14 at 1 μl, washed and then incubated for I hour with a secondary anti-mouse Fc antibody conjugated to horseradish peroxidase ("Horse Radish peroxidase") (sold by the company Sigma). Several washes were carried out before revelation using the ECL detection kit sold by the company Amersham.
Pour l'analyse de la séquence d'aminoacides, la protéine ESM- 1 purifiée a été soumise à une électrophorèse sur un gel SDS-PAGE, puis électrotransférée sur une membrane de difluorure de polyvinylidène (PVDF) commercialisée par la Société MILLIPORE, puis colorée à l'aide de 0,1% de bleu de coomassie. La bande protéine a 50 kD a été excisée de la membrane et la séquence N-terminale a été effectuée par dégradation d'EDMAN automatisée sur un sequenceur de protéine du type ABI 473A.For the analysis of the amino acid sequence, the purified ESM-1 protein was subjected to electrophoresis on an SDS-PAGE gel, then electrotransfered onto a polyvinylidene difluoride (PVDF) membrane sold by the company Millipore, then stained. using 0.1% coomassie blue. The protein band at 50 kD was excised from the membrane and the N-terminal sequence was carried out by degradation of automated EDMAN on a protein sequencer of the ABI 473A type.
A.6 Digestion du corps peptidique de ESM-1 par la proteinase K Afin de déterminer la taille du glycosaminoglycane, la protéine ESM-1 purifiée a été digérée avec la proteinase K avec un rapport enzyme:ESM-1 égal à 1 :50 (poids:poids) dans un tampon Tris 10mM, pH8, en présence ou en absence de 0,1% SDS à 56°C pendant 3 heures. Une quantité 10 supérieure de sérum albumine bovine (BSA) à la protéine ESM-1 a été digérée par la proteinase K afin de vérifier sa dégradation complète. Les échantillons ont été analysés sur un gel de 12 % SDS-PAGE, suivi par une coloration au bleu de coomassie et au bleu alcian.A.6 Digestion of the peptide body of ESM-1 by proteinase K In order to determine the size of the glycosaminoglycan, the purified ESM-1 protein was digested with proteinase K with an enzyme: ESM-1 ratio equal to 1: 50 (weight: weight) in a 10 mM Tris buffer, pH8, in the presence or in the absence of 0.1% SDS at 56 ° C for 3 hours. A greater amount of bovine serum albumin (BSA) than the protein ESM-1 was digested by proteinase K in order to verify its complete degradation. The samples were analyzed on a 12% SDS-PAGE gel, followed by staining with coomassie blue and alkian blue.
A.7 Digestion de ESM-1 par les chondroïtinases ABC. B. AC et CA.7 Digestion of ESM-1 with ABC chondroitinases. B. AC and C
Afin d'analyser la nature de la substitution de glycosaminoglycane, la protéine ESM-1 purifiée a été digérée avec plusieurs chondroïtinases: chondroïtinases ABC (0,5 unité/ mg dans un tampon 100 mM TrisHCI, pH 8, 30 mM d'acétate de sodium, pH 5,2 à 37°C pendant 45 min), chondroïtinase B (200 unités/mg dans un tampon 20 mM Tris-HCI, 50 mM Nacl, 4 mM CaCI2, 0,01% BSA, pH 7,5 à 25°C pendant deux heures), des chondroïtinase AC (une unité par ml dans un tampon 250 mM Tris Hcl, 75 mM acétate de sodium, pH 7,3 à 37°C pendant deux heures) des chondroïtinases C (80-120 unités/ml dans un tampon 50 mM Tris HCI , pH 8 à 25°C pendant 3 heures). Les échantillons ont été analysés par immuno-empreinte.In order to analyze the nature of the glycosaminoglycan substitution, the purified ESM-1 protein was digested with several chondroitinases: ABC chondroitinases (0.5 unit / mg in 100 mM TrisHCI buffer, pH 8, 30 mM acetate sodium, pH 5.2 at 37 ° C for 45 min), chondroitinase B (200 units / mg in 20 mM Tris-HCl buffer, 50 mM Nacl, 4 mM CaCl 2 , 0.01% BSA, pH 7.5 at 25 ° C for two hours), chondroitinase AC (one unit per ml in 250 mM Tris Hcl buffer, 75 mM sodium acetate, pH 7.3 at 37 ° C for two hours) chondroitinases C (80-120 units / ml in 50 mM Tris HCl buffer, pH 8 at 25 ° C for 3 hours). The samples were analyzed by immunoblotting.
A.8 Activité anti-coagulante.A.8 Anticoagulant activity.
Le plasma témoin pauvre en plaquette (PPP) a été préparé à partir de sang en présence de l'anticoagulant citrate de sodium (30mM), par centrifugation à 2500 g pendant 15 min. Tous les réactifs sont commercialisés par la Société STAGO Diagnostica (France). Trois paramètres ont été évalués, en ajoutant la protéine ESM-1 , du tampon ou de l'héparine au plasma pauvre en plaquette: a) APTT (pour « Activated Partialo Thromboplastin Time »): ce paramètre explore la voie intrinsèque de la coagulation sanguine (FI, Fil, FV, FVIII, FIX, FX, FXI, FXII). Le déficit ou l'inhibition de l'un de ces facteurs augmente le temps de coagulation du mélange réactif PPP, céphaline, activateur, CACI2. b) TCT (pour « Thrombin Clotting Time »): ce paramètre est analysé sur un mélange de plasma pauvre en plaquette (PPP) en présence de thrombine. Avec une concentration standard de thrombine, le temps de coagulation du plasma est constant. Des défauts dans la formation de fibrine induisent une augmentation du temps de coagulation. c) activité anti-Xa: l'activité anti-Xa de l'héparine ou d'autres inhibiteurs agissant sur le facteur FXa est détectée par un test compétitif. L'échantillon étudié (PPP + ESM-1 , + tampon ou + héparine) est mélangé avec le facteur Fxa et un substrat chromogène spécifique du facteur Fxa. La coloration finale est inversement proportionnelle à la concentration d'inhibiteur.The control plasma poor in platelets (PPP) was prepared from blood in the presence of the anticoagulant sodium citrate (30 mM), by centrifugation at 2500 g for 15 min. All reagents are marketed by STAGO Diagnostica (France). Three parameters were evaluated, by adding the ESM-1 protein, buffer or heparin to the plasma poor in platelets: a) APTT (for “Activated Partialo Thromboplastin Time”): this parameter explores the intrinsic pathway of blood coagulation (FI, Wire, FV, FVIII, FIX, FX, FXI, FXII). The deficit or inhibition of one of these factors increases the coagulation time of the PPP reactive mixture, cephalin, activator, CACI 2 . b) TCT (for “Thrombin Clotting Time”): this parameter is analyzed on a mixture of plasma poor in platelets (PPP) in the presence of thrombin. With a standard concentration of thrombin, the plasma coagulation time is constant. Defects in fibrin formation induce an increase in clotting time. c) anti-Xa activity: the anti-Xa activity of heparin or of other inhibitors acting on the factor FXa is detected by a competitive test. The studied sample (PPP + ESM-1, + buffer or + heparin) is mixed with factor Fxa and a chromogenic substrate specific for factor Fxa. The final coloration is inversely proportional to the concentration of inhibitor.
A.9 Test de génération de la thrombineA.9 Thrombin generation test
Ce test global sensible peut détecter des défauts plasmatiques ou dans les plaquettes induisant un retard ou une réduction dans la génération de thrombine. A partir de sang, un plasma riche en plaquettes (PRP) a été préparé à partir de sang en présence de citrate de sodium par centrifugation à 150 g pendant 10 min . Le test de génération de la thrombine a été réalisé, pour chacun des sujets, dans des échantillons en l'absence de ESM-1 , avec de l'héparinate de calcium non fractionnée (0,5 Ul de anti-Xa/ml) ou avec 0,2mg/ml, 0,5 mg/ml et 1 mg/ml de ESM-1 (concentration finale).This sensitive global test can detect plasma or platelet defects inducing a delay or a reduction in the generation of thrombin. From blood, a platelet rich plasma (PRP) was prepared from blood in the presence of sodium citrate by centrifugation at 150 g for 10 min. The thrombin generation test was carried out, for each of the subjects, in samples in the absence of ESM-1, with unfractionated calcium heparinate (0.5 IU of anti-Xa / ml) or with 0.2mg / ml, 0.5 mg / ml and 1 mg / ml ESM-1 (final concentration).
La protéine ESM-1 a été ajoutée 10 min avant le test. A 37°C, 1 ml de plasma a été mélangé avec 1 ml de CaCI2 et le chronomètre a été déclenché. Des fractions aliquotes de 0,1 ml ont été reprises du mélange réactif chaque minute pendant 1 min.The ESM-1 protein was added 10 min before the test. At 37 ° C, 1 ml of plasma was mixed with 1 ml of CaCl 2 and the stopwatch was started. 0.1 ml aliquots were taken up from the reaction mixture every minute for 1 min.
Le caillot formé dans le mélange réactif est régulièrement éliminé. Les fractions aliquotes ont été mélangées avec 0,2 ml de fibrinogène (Sigma, 4/1000 dans du tampon Owren à 37°C et le temps de coagulation a été mesurée pour chacune des fractions aliquotes). La thrombine formée dans le mélange réactif agit sur les forminogènes, induisant la formation de fibrine. L'activité de coagulation était maximale entre 4 et 8 min puis a diminué du fait de la neutralisation de la thrombine par l'anti-thrombine.The clot formed in the reaction mixture is regularly removed. The aliquots were mixed with 0.2 ml of fibrinogen (Sigma, 4/1000 in Owren buffer at 37 ° C and the coagulation time was measured for each of the aliquots). The thrombin formed in the reaction mixture acts on the forminogens, inducing the formation of fibrin. The coagulation activity was maximum between 4 and 8 min and then decreased due to the neutralization of the thrombin by the anti-thrombin.
A.10 Chromatographie analytique par filtration sur gelA.10 Analytical chromatography by gel filtration
50 μg de ESM-1 glycosylée (ESM/WT) purifiée et de ESM-1 non glycosylée (ESM/S137A) purifiée dans du tampon 50 mM Tris, pH 8,5, 0,5 MNAcl ont été séparées par chromatographie liquide sur une colonne Superdex 200 (pour ESM/WT) ou Superdex 75 (pour ESM/S137A) commercialisée par Pharmacia, en utilisant le système de chromatographie Biorad Biologie Chromatography System avec un débit de 1 ml/min. Comme standard, on a utilisé le kit de calibration de haut et bas poids moléculaire (Pharmacia Biotech) suivant : ribonucléase A (de pancréas bovin, 13,7 kD), ovalbumine (43 kD) albumine (sérum bovin, 67 kd), aldolase (muscle de lapin, 158 kD), ferritine (rate de cheval, 440 kD), thiroglobuline (thyroïde bovine, 669 kD). Les standards de poids moléculaire ont été séparés en utilisant un tampon identique à celui utilisé pour les protéines ESM-1 et la séparation a été réalisée immédiatement après la séparation des protéines ESM/WT et ESM/S137A. Le temps d'élution des protéines standard a été utilisé pour construire une courbe linéaire standard, Kav = f(log MR) afin de déterminer le poids moléculaire apparent respectif des protéines ESM/WT et ESM/S137A.50 μg of purified ESM-1 glycosylated (ESM / WT) and non-glycosylated ESM-1 (ESM / S137A) purified in 50 mM Tris buffer, pH 8.5, 0.5 MNAcl were separated by liquid chromatography on a Superdex 200 column (for ESM / WT) or Superdex 75 column (for ESM / S137A) sold by Pharmacia, using the Biorad Biologie Chromatography System with a flow rate of 1 ml / min. As standard, the following high and low molecular weight calibration kit (Pharmacia Biotech) was used: ribonuclease A (from bovine pancreas, 13.7 kD), ovalbumin (43 kD) albumin (bovine serum, 67 kd), aldolase (rabbit muscle, 158 kD), ferritin (horse spleen, 440 kD), thiroglobulin (bovine thyroid, 669 kD). The molecular weight standards were separated using a buffer identical to that used for the ESM-1 proteins and the separation was carried out immediately after the separation of the ESM / WT and ESM / S137A proteins. The standard protein elution time was used to construct a standard linear curve, Kav = f (log MR) to determine the respective apparent molecular weights of the ESM / WT and ESM / S137A proteins.
Des fractions de 1 ml ont été recueillies et la protéine ESM-1 a été détectée à l'aide d'un test immunoenzymatique (ELISA) spécifique.1 ml fractions were collected and the ESM-1 protein was detected using a specific enzyme-linked immunosorbent assay (ELISA).
B. RESULTATSB. RESULTS
B.1 Modifications post-traductionnelles de la forme sécrétée de la protéine ESM-1 produite par les cellules endothéliales et par des lignées cellulaires établies. Afin de déterminer si ESM-1 était maturée comme une molécule sécrétée, comme cela a été suggéré par la présence d'une séquence d'aminoacides N-terminale prédite comme un peptide signal, la protéine ESM-1 a été purifiée à partir de la lignée cellulaire 293-ESM. La séquence N-terminale de la forme de 50 kD a indiqué que le peptide signal de 19 acides aminés était clivé au site prédit, résultant en un polypeptide ESM-1 mature de 165 acides aminés commençant au résidu tryptophane en position 20 de la séquence SEQ ID N°1 , la séquence N-terminale étant « WSNNYAVD-P ». ESM-1 a été immunoprécipité à partir des surnageants de cultures cellulaires des cellules HUVEC, SV1 , 293-ESM et CHO-ESM, puis analysés par immuno-empreinte.B.1 Post-translational modifications of the secreted form of the ESM-1 protein produced by endothelial cells and by established cell lines. In order to determine if ESM-1 was matured as a secreted molecule, as suggested by the presence of an N-terminal amino acid sequence predicted as a signal peptide, the protein ESM-1 was purified from the cell line 293-ESM. The N-terminal sequence of the 50 kD form indicated that the signal peptide of 19 amino acids was cleaved at the predicted site, resulting in a mature ESM-1 polypeptide of 165 amino acids starting at the tryptophan residue in position 20 of the sequence SEQ ID No. 1, the N-terminal sequence being "WSNNYAVD-P". ESM-1 was immunoprecipitated from the cell culture supernatants of HUVEC, SV1, 293-ESM and CHO-ESM cells, then analyzed by immunoblotting.
Dans les surnageants des cellules HUVEC, il a été montré précédemment que ESM-1 migrait sous la forme d'une bande diffuse à environ 50 kD.In the supernatants of HUVEC cells, it was previously shown that ESM-1 migrated in the form of a diffuse band at around 50 kD.
Une bande similaire en taille a été observée avec les surnageants des cellules SV1 , 293-ESM et CHO-ESM (figure 1A).A similar band in size was observed with the supernatants of cells SV1, 293-ESM and CHO-ESM (FIG. 1A).
Le poids moléculaire retrouvé est plus important que le poids moléculaire prédit. Ce résultat a suggéré que la forme sécrétée de ESM- 1 avait subi des modifications post-traductionnelles. Le fait que la protéine ESM-1 purifiée était mieux colorée sur le gel SDS-PAGE avec le bleu alcian qu'avec le bleu de coomassie a suggéré que ESM-1 était glycosylée (figures 1B, 1C) plutôt que oligomérisée au travers de pont disulfure, du fait que des conditions réductrices ne modifient pas le poids moléculaire apparent de ESM-1.The molecular weight found is more important than the predicted molecular weight. This result suggested that the secreted form of ESM-1 had undergone post-translational modifications. The fact that the purified ESM-1 protein was better stained on the SDS-PAGE gel with alkian blue than with coomassie blue suggested that ESM-1 was glycosylated (FIGS. 1B, 1C) rather than oligomerized across the bridge disulfide, since reducing conditions do not change the apparent molecular weight of ESM-1.
B.2. Le résidu serine en position 137 (SEQ ID N°1) est le site de O- glycosylation de ESM-1.B.2. The serine residue at position 137 (SEQ ID No. 1) is the O-glycosylation site of ESM-1.
Une analyse par ordinateur des sites de glycosylation potentielle a permis d'identifier trois sites putatifs de O-glycosylation, respectivement pour la serine en position 16, sur la thréonine en positionA computer analysis of the potential glycosylation sites made it possible to identify three putative sites of O-glycosylation, respectively for the serine in position 16, on the threonine in position
120 et sur la serine en position 127, mais aucun site de N-glycosylation.120 and on the serine at position 127, but no N-glycosylation site.
Le résidu thréonine en position 120 et le résidu serine en position 137 ont été mutés et remplacés par un résidu alanine. Ces mutants ont été exprimés de manière transitoire dans les cellules 293.The threonine residue at position 120 and the serine residue at position 137 have been mutated and replaced by an alanine residue. These mutants were transiently expressed in 293 cells.
La protéine ESM-1 a ensuite été immunoprécipitée à partir des lysats cellulaires et des surnageants de culture, et analysés par immuno- empreinte.The ESM-1 protein was then immunoprecipitated from cell lysates and culture supernatants, and analyzed by immunoblotting.
La protéine ESM/T120A migre à 50 kD, à une position similaire au poids moléculaire apparent de la forme sauvage de ESM-1 (ESM/WT), comme cela est montré à la figure 2A.The ESM / T120A protein migrates at 50 kD, at a position similar to the apparent molecular weight of the wild form of ESM-1 (ESM / WT), as shown in Figure 2A.
Au contraire, la protéine ESM/S137A migre à 22 kD correspondant à la forme intra-cellulaire de ESM-1 (figure 2A) , un poids moléculaire compatible avec le poids moléculaire prédit.de ESM-1.In contrast, the ESM / S137A protein migrates to 22 kD corresponding to the intracellular form of ESM-1 (Figure 2A), a molecular weight compatible with the predicted molecular weight of ESM-1.
Les immunoprécipitations réalisées à partir des cellules COS et CHO transfectees transitoirement ont donné les mêmes résultats, indiquant que seul le résidu serine en position 137 constituait un site de glycoconjuguaison dans tous les modèles cellulaires étudiés.Immunoprecipitations carried out from transiently transfected COS and CHO cells gave the same results, indicating that only the serine residue at position 137 constituted a glycoconjugation site in all the cell models studied.
Afin de déterminer la longueur du glycosaminoglycane (GAG) de ESM-1, le corps peptidique de ESM-1 a été complètement digéré par la proteinase K.In order to determine the length of the glycosaminoglycan (GAG) of ESM-1, the peptide body of ESM-1 was completely digested by proteinase K.
Le traitement par la proteinase K induit un changement dans le poids moléculaire de 50 kD à 25-30 kD (figure 2B). Ces résultats montrent que la bande de poids moléculaire apparent à 50. kD est compatible avec la présence d'un polypeptide de 22 kD qui est glycoconjuguée sur la serine en position 137 par une chaîne GAG d'une taille moyenne de 25-30 kD.Proteinase K treatment induces a change in molecular weight from 50 kD to 25-30 kD (Figure 2B). These results show that the band of apparent molecular weight at 50. kD is compatible with the presence of a 22 kD polypeptide which is glycoconjugated on the serine at position 137 by a GAG chain with an average size of 25-30 kD.
B.3 La chaîne GAG DE ESM-1 est sensible à la chondroïtinase ABCB.3 The GAG chain of ESM-1 is sensitive to chondroitinase ABC
Afin de caractériser la chaîne GAG de ESM-1 , la protéine ESM-In order to characterize the GAG chain of ESM-1, the protein ESM-
1 a été tout d'abord digérée par la chondroïtinase ABC. Le traitement par la chondroïtinase ABC a réduit le poids moléculaire de la protéine ESM-1 sécrétée à 22 kD (figure 3A), suggérant que le carbohydrate de ESM-1 est une chaîne du type chondroïtine.1 was first digested with chondroitinase ABC. Treatment with chondroitinase ABC reduced the molecular weight of the secreted protein ESM-1 to 22 kD (Figure 3A), suggesting that the carbohydrate of ESM-1 is a chain of the chondroitin type.
Le profil est similaire avec la protéine ESM-1 purifiée à partir des cellules 293-ESM ainsi qu'à partir de la lignée cellulaire endothéliale humaine SVI. Du fait que la protéine ESM-1 circule dans le sang, non a également étudié le comportement de la protéine ESM-1 purifiée à partir de plasma humain. Les résultats permettent d'observer une bande unique principale de 50 kD, qui a un poids moléculaire de 22 kD après traitement avec la chondroïtinase ABC, comme pour l'ensemble des autres lignées cellulaires étudiées (figure 3A). Ainsi, la protéine ESM-1 est un protéoglycane soluble contenant une chaîne unique de sulfate de chondroïtine.The profile is similar with the protein ESM-1 purified from 293-ESM cells as well as from the human endothelial cell line SVI. Because the protein ESM-1 circulates in the blood, not a also studied the behavior of the protein ESM-1 purified from human plasma. The results make it possible to observe a single main band of 50 kD, which has a molecular weight of 22 kD after treatment with chondroitinase ABC, as for all the other cell lines studied (FIG. 3A). Thus, the ESM-1 protein is a soluble proteoglycan containing a single chain of chondroitin sulfate.
B.4 La chaîne GAG de ESM-1 est une chaîne hétérogène de chondroïtine/dermatane sulfate.B.4 The GAG chain of ESM-1 is a heterogeneous chain of chondroitin / dermatan sulfate.
Afin de mieux déterminer le type de motif dit « saccharidiques » qui constitue la chaîne GAG de ESM-1 , on a utilisé plusieurs enzymes spécifiques, telles que les chondroïtinases B, AC et C. Le traitement à la chondroïtinase B réduit le poids moléculaire apparent de 50 kD à 22 kD (figure 3B).In order to better determine the type of motif called "saccharides" which constitutes the GAG chain of ESM-1, several specific enzymes were used, such as chondroitinases B, AC and C. Treatment with chondroitinase B reduces the apparent molecular weight from 50 kD to 22 kD (Figure 3B).
Un profil similaire a été observé après traitement de l'ESM-1 par les chondroïtinases AC et C (figures 3C, D) .A similar profile was observed after treatment of ESM-1 with chondroitinases AC and C (Figures 3C, D).
Ces traitements enzymatiques différents indiquent que la chaîne GAG de ESM-1 contient différents motifs composites contenant un type de sucre aminé, la N-acétylgalactosamine, couplée à l'acide iduronique ou glucuronique différemment sulfatée.These different enzymatic treatments indicate that the GAG chain of ESM-1 contains different composite units containing a type of amino sugar, N-acetylgalactosamine, coupled to differently sulphated iduronic or glucuronic acid.
Ces différents motifs seraient pris en alternance dans la chaîne, et seraient présents au début de la chaîne, à proximité des dissacharides sulfatés N-terminaux qui persistent sur le corps protéique après digestion à la chondroïtinase, du fait que tous les traitements à la chondroïtinase conduisent aux mêmes poids moléculaire apparents réduits de 22 kD.These different motifs would be taken alternately in the chain, and would be present at the start of the chain, near the N-terminal sulfated dissacharides which persist on the protein body after chondroitinase digestion, because all chondroitinase treatments lead at the same apparent molecular weights reduced by 22 kD.
B.5. Activité biologique du protéoglycane soluble ESM-1 sur la coagulationB.5. Biological activity of the soluble proteoglycan ESM-1 on coagulation
Du fait que la protéine ESM-1 est sécrétée comme un protéoglycane du type chondroïtine/dermatane sulfate par les cellules endothéliales et du fait que le sulfate de dermatane présente des effets sur la génération de thrombine in vitro DELORME et al., (1996) et sur la coagulation, l'activité anticoagulante potentielle de ESM-1 a été vérifiée sur les paramètres APTT, TCT, l'activité anti-Xa et sur la génération de thrombine.Because the ESM-1 protein is secreted as a proteoglycan of the chondroitin / dermatan sulfate type by endothelial cells and because dermatan sulfate has effects on the generation of thrombin in vitro DELORME et al., (1996) and on coagulation, the potential anticoagulant activity of ESM-1 has been verified on the parameters APTT, TCT, anti-Xa activity and on the generation of thrombin.
Les résultats sont représentés dans le tableau 1 ci-après.The results are shown in Table 1 below.
TABLEAU 1 Activité biologique du protéoglycane ESM-1 sur la coagulationTABLE 1 Biological activity of the ESM-1 proteoglycan on coagulation
Les résultats du tableau 1 montrent que la protéine ESM-1 aux différentes doses importantes de 0,2 mg/ml à 1 mg/ml ne modifient pas les différents paramètres testés.The results in Table 1 show that the ESM-1 protein at the various large doses of 0.2 mg / ml to 1 mg / ml does not modify the various parameters tested.
Les paramètres APTT, TCT et activité anti-Xz sont similaires pour le plasma pauvre en plaquettes (PPP) avec le tampon ou avec la protéine ESM-1. Dans les témoins positifs, les activités APTT, TCT et anti-Xa sont plus grandes pour le PPP en présence d'héparine.The APTT, TCT and anti-Xz activity parameters are similar for platelet-poor plasma (PPP) with the buffer or with the protein ESM-1. In the positive controls, the APTT, TCT and anti-Xa activities are greater for the PPP in the presence of heparin.
De plus, la protéine ESM-1 n'a pas d'effet inhibiteur sur le test de génération de la thrombine: on observe aucune différence selon les concentrations de 0,2 mg/ml, 0,5 mg/ml et 1 mg/ml de ESM-1 par rapport au témoin tampon, alors que l'héparine induit un retard dans la formation de thrombine (figure 4).In addition, the ESM-1 protein has no inhibitory effect on the thrombin generation test: there is no difference according to the concentrations of 0.2 mg / ml, 0.5 mg / ml and 1 mg / ml of ESM-1 relative to the buffer control, while heparin induces a delay in the formation of thrombin (FIG. 4).
EXEMPLE 2:EXAMPLE 2:
Effet de la protéine ESM-1 sur l'activité mitoqène du facteur HGF/SFEffect of the protein ESM-1 on the mitoqene activity of factor HGF / SF
A. Matériels et MéthodesA. Materials and Methods
L'activité de stimulation de la prolifération a été déterminée en mesurant l'incorporation de 3H thymidine par les cellules 293. Les cellules 293 ont été ensemencées à la concentration de 1 x 104 cellules par puits dans des microplaques à 96 puits du type TPP et ont été maintenues pendant 24 heures dans le milieu de culture DMEM additionnées de transférine et d'insuline. Le HGF/SF recombinant humain a été dilué dans du PBS contenant 0,1% de sérum albumine bovine et ajouté par l'eau de 3 puits identiques afin d'obtenir une concentration finale de 50 ng/ml.The proliferation-stimulating activity was determined by measuring the incorporation of 3 H thymidine by the 293 cells. The 293 cells were seeded at the concentration of 1 × 10 4 cells per well in 96-well microplates of the TPP type and were maintained for 24 hours in the DMEM culture medium supplemented with transferin and insulin. The recombinant human HGF / SF was diluted in PBS containing 0.1% bovine serum albumin and added with water from 3 identical wells in order to obtain a final concentration of 50 ng / ml.
Les protéines recombinantes ESM/WT, ESM/S137A, la chaîne GAG purifiée dérivée de ESM-1 et la décorine ont été ajoutées seules ou en association avec le facteur HGF/SF à des doses allant de 1 ng/ml à 2,5 μg/ml, simultanément à l'addition de HGF/SF.The recombinant proteins ESM / WT, ESM / S137A, the purified GAG chain derived from ESM-1 and decorin were added alone or in combination with the factor HGF / SF at doses ranging from 1 ng / ml to 2.5 μg / ml, simultaneously with the addition of HGF / SF.
Après 96 heures de culture, les cellules ont été incubées avec 0,5 μCi de 3H thymidine par puits pendant 16 heures et l'incorporation de 3H thymidine a été déterminée à l'aide d'un compteur à scintillation du type Topcount Microplate Scintillation Counter (Packard).After 96 hours of culture, the cells were incubated with 0.5 μCi of 3 H thymidine per well for 16 hours and the incorporation of 3 H thymidine was determined using a scintillation counter of the Topcount Microplate type. Scintillation Counter (Packard).
Les tests ont été réalisés sur des lots de trois puits identiques.The tests were carried out on batches of three identical wells.
La viabilité cellulaire a été mesurée à l'aide du test de réduction de MTT.Cell viability was measured using the MTT reduction test.
B. RESULTATSB. RESULTS
L'effet de la protéine ESM-1 sur l'activité du facteur HGF/SF a été étudié.The effect of the protein ESM-1 on the activity of factor HGF / SF has been studied.
L'incorporation de 3H-thymidine par les cellules 293 a été mesurée en présence de HGFSF à 50 ng/ml seule ou en association avec différentes quantités de ESM/WT.The incorporation of 3 H-thymidine by the 293 cells was measured in the presence of HGFSF at 50 ng / ml alone or in combination with different amounts of ESM / WT.
Dans un premier lot d'expériences, on a observé que le HGF/SF seul à 50 ng/ml induisait une prolifération des cellules 293 à un niveau égal à environ 45% de la prolifération induite par le sérum, alors que la protéine ESM/WT seule ne stimulait pas la prolifération des cellules 293.In a first batch of experiments, it was observed that HGF / SF alone at 50 ng / ml induces proliferation of 293 cells at a level equal to approximately 45% of the proliferation induced by serum, while the protein ESM / WT alone did not stimulate the proliferation of 293 cells.
En revanche, lorsqu'elle est combinée au facteur HGF/SF, la protéine ESM/WT augmente considérablement la prolifération des cellules 293 induites par HGF/SF avec une augmentation de 162,3%, lorsque' la protéine est testée à la concentration de 2,5 μg/ml (figure 5). Cet effet d'augmentation de la protéine ESM-1 sur l'activité HGF/SF est dépendant de la dose ESM-1 et commence a être significative à la dose de 10 ng/ml (figure 6).On the other hand, when combined with the HGF / SF factor, the ESM / WT protein considerably increases the proliferation of HGF / SF-induced 293 cells with an increase of 162.3%, when the protein is tested at the concentration of 2.5 μg / ml (Figure 5). This effect of increasing the protein ESM-1 on the HGF / SF activity is dependent on the dose ESM-1 and begins to be significant at the dose of 10 ng / ml (FIG. 6).
De plus, on a comparé l'effet de la protéine ESM/WT à l'effet de la décorine, un autre protéoglycane du type chondroïtine sulfate/dermatane sulfate, sur l'activité mitogène du facteur HGF/SF. Au contraire de la protéine ESM/WT, la décorine ne présente aucune activité d'augmentation de la prolifération des cellules 293 induites par le facteur HGF/SF (figures 5, 6). Ces résultats indiquent que la protéine ESM-1 a un effet spécifique sur l'activité mitogène du facteur HGF/SF.In addition, the effect of the protein ESM / WT was compared with the effect of decorin, another proteoglycan of the chondroitin sulfate / dermatan sulfate type, on the mitogenic activity of the HGF / SF factor. Unlike the protein ESM / WT, decorin does not show any activity for increasing the proliferation of 293 cells induced by the HGF / SF factor (FIGS. 5, 6). These results indicate that the protein ESM-1 has a specific effect on the mitogenic activity of factor HGF / SF.
Afin d'examiner l'implication respective du corps protéique de ESM-1 et de la chaîne GAG sur l'activité d'augmentation de l'effet mitogène, on a mesuré l'incorporation de 3H-thymidine par les cellules 293 en présence de HGF/SF additionnées de différentes concentrations de ESM/S137A non glycosylée et de la chaîne GAG dérivée de ESM-1.In order to examine the respective involvement of the protein body of ESM-1 and of the GAG chain on the activity of increasing the mitogenic effect, the incorporation of 3 H-thymidine by the 293 cells was measured. of HGF / SF supplemented with different concentrations of non-glycosylated ESM / S137A and of the GAG chain derived from ESM-1.
La forme non glycosylée de ESM-1 est incapable d'induire une prolifération des cellules 293, que ce soit en présence ou en l'absence du facteur HGFSF (figure 5), même lorsque elle est utilisée à haute concentration.The non-glycosylated form of ESM-1 is incapable of inducing proliferation of 293 cells, whether in the presence or in the absence of the factor HGFSF (FIG. 5), even when it is used in high concentration.
Au contraire, la chaîne GAG purifiée à partir de ESM-1 augmente considérablement la prolifération des cellules 293 induites par le facteur HGF/SF, avec un facteur d'augmentation proche de 96,6%, comparée au facteur HGF/SF seul (figure 5). L'effet pro-mitogène de la chaîne GAG est moindre que celui observé avec la forme sauvage de la protéine ESM-1 , mais cet effet est néanmoins dépendant de la dose de chaîne GAG ajoutée (figure 6).On the contrary, the GAG chain purified from ESM-1 considerably increases the proliferation of 293 cells induced by the HGF / SF factor, with an increase factor close to 96.6%, compared to the HGF / SF factor alone (FIG. 5). The pro-mitogenic effect of the GAG chain is less than that observed with the wild form of the protein ESM-1, but this effect is nevertheless dependent on the dose of GAG chain added (FIG. 6).
Les résultats détaillés ci-dessus démontrent clairement que la protéine ESM/WT augmente la prolifération des cellules 293 induites par le facteur HGF/SF et que cette activité pro-mitogène est spécifique et due à la chaîne GAG du type chondroïtine sulfate/dermatane sulfate de ESM-1.The results detailed above clearly demonstrate that the ESM / WT protein increases the proliferation of 293 cells induced by the HGF / SF factor and that this pro-mitogenic activity is specific and due to the GAG chain of the chondroitin sulfate / dermatan sulfate type. ESM-1.
De manière générale, le facteur HGF/SF est exprimé durant les critiques périodes précoces de l'organogénèse humaine de 6 à 13 semaines de gestation. Les organes qui expriment le gène de HGF/SF sont notamment le foie, le rein métanéphrique, l'intestin et le poumon, chacun de ces organes se développant par interaction inductive entre le mesenchyme et l'épithélium. De plus, le facteur HGF/SF est un facteur important dans la dysplasie multicystique rénale humaine (TAKAYAMA et al., 1997) ainsi que dans l'apparition de malformation et d'hyperprolifération dans les tubules. Les résultats présentés ci-dessus indiquent que la protéine ESM-1 augmente significativement la prolifération des cellules du rein embryonnaire en présence de HGF/SF alors que la forme non glycosylée et de la protéine ESM-1 n'a pas d'effet. De plus, la chaîne GAG isolée à partir de la protéine ESM-1 est capable \ de mimer les effets de la protéine glycosylée ESM/WT. Ces résultats démontrent clairement que l'activité biologique de ESM-1 sur la fonction du facteur HGF/SF est médiée principalement par sa chaîne GAG. On peut noter que la décorine, un autre protéoglycane du type chondroïtine sulfate/dermatane sulfate sécrété par les cellules endothéliales et qui est capable de se fixer sur le facteur HGF/SF (CELLA et al. , 1992) n'a pas d'effet sur l'activité de HGFSF. Ces comparaisons indiquent une spécificité d'action de la protéine ESM-1 sur l'activité du facteur HGF/SF requérant une composition de la chaîne GAG différente de la chaîne GAG des protéoglycanes appartenant à la famille des protéoglycanes à petites répétitions riches en leucine (« small leucine reach repeats »).In general, the HGF / SF factor is expressed during the critical early periods of human organogenesis of 6 to 13 weeks of gestation. Organs that express the HGF / SF gene are notably the liver, the metanephric kidney, the intestine and the lung, each of these organs developing by inductive interaction between the mesenchyme and the epithelium. In addition, the HGF / SF factor is an important factor in human renal multicystic dysplasia (TAKAYAMA et al., 1997) as well as in the appearance of malformation and hyperproliferation in the tubules. The results presented above indicate that the ESM-1 protein significantly increases the proliferation of embryonic kidney cells in the presence of HGF / SF while the non-glycosylated form and of the ESM-1 protein has no effect. In addition, the GAG chain isolated from the ESM-1 protein is capable of mimicking the effects of the glycosylated protein ESM / WT. These results clearly demonstrate that the biological activity of ESM-1 on the function of the HGF / SF factor is mainly mediated by its GAG chain. It may be noted that decorin, another proteoglycan of the chondroitin sulfate / dermatan sulfate type secreted by endothelial cells and which is capable of binding to the factor HGF / SF (CELLA et al., 1992) has no effect on the activity of HGFSF. These comparisons indicate a specificity of action of the protein ESM-1 on the activity of factor HGF / SF requiring a composition of the GAG chain different from the GAG chain of proteoglycans belonging to the family of proteoglycans with small repetitions rich in leucine ( "Small leucine reach repeats").
Dans le rein, la protéine ESM-1 est détectée sélectivement dans les tubules distaux, le résultat qui peut être associé à l'observation de la localisation préférentielle du facteur HGF/SF dans la même partie, du néphron dans des situations de dysplasies multicystique rénale humaine (WEIDNER et al., 1993). Ces résultats indiquent une application de la protéine ESM-1 dans des troubles pathologiques dépendant du facteur HGF/SF, qui a également été montré comme étant associé dans le développement de cancers du sein (RAHIMI et al., 1998), du rein (NATALI et al, (1996)) et du poumon (OTSUKA et al. , 1998) mais aussi dans les mélanomes malins (SIEGFRIED et al., 1998). Ainsi, le facteur HGF/SF est susceptible de favoriser l'extension de l'hyperplasie et d'engendrer des cellules avec un phénotype invasif. La protéine ESM-1 est susceptible d'être impliquée dans ces phénomènes d'activités mitogènes dérégulés du facteur HGF/SF. EXEMPLE 3 :In the kidney, the protein ESM-1 is selectively detected in the distal tubules, the result which can be associated with the observation of the preferential localization of the HGF / SF factor in the same part, of the nephron in situations of renal multicystic dysplasia human (WEIDNER et al., 1993). These results indicate an application of the protein ESM-1 in pathological disorders dependent on the factor HGF / SF, which has also been shown to be associated in the development of cancers of the breast (RAHIMI et al., 1998), of the kidney (NATALI et al, (1996)) and lung (OTSUKA et al., 1998) but also in malignant melanomas (SIEGFRIED et al., 1998). Thus, the HGF / SF factor is likely to promote the extension of hyperplasia and to generate cells with an invasive phenotype. The ESM-1 protein is likely to be involved in these phenomena of deregulated mitogenic activities of the HGF / SF factor. EXAMPLE 3:
Préparation d'un composé antagoniste de la protéine ESM-1 du type anticorps.Preparation of an antagonist compound of the antibody-type protein ESM-1.
Afin d'obtenir des anticorps monoclonaux anti-ESM-1 dirigés contre la région N-terminale de la protéine ESM-1 riche en résidus cystéine, la forme native de la protéine ESM-1 produite par la lignée cellulaire CHO transfectée par un vecteur d'expression contenant un insert d'ADN codant pour la protéine ESM-1 a été purifiée.In order to obtain anti-ESM-1 monoclonal antibodies directed against the N-terminal region of the ESM-1 protein rich in cysteine residues, the native form of the ESM-1 protein produced by the CHO cell line transfected with a vector d the expression containing a DNA insert coding for the protein ESM-1 has been purified.
L'ADNc de ESM-1 est inséré dans le vecteur d'expression eucaryote pcDNA3 (In vitrogen) puis transfecté dans les cellules CHO avec la lipofectamine (GIBCO) selon les recommandations du fabricant. 48 heures après la transfection les cellules sont repiquées en présence d'un agent de sélection (G418, Gibco) à la dose de 1000 microgramme/ml). Après deux semaines de sélection, les cellules CHO résistantes au G418 sont données par dilution limite. Des clones exprimant ESM-1 sont ensuite sélectionnés et appelés CHO-ESM (déposé à la CNCM). Pour la production, les cellules CHO-ESM sont cultivées en suspension dans un milieu conditionné sans sérum de veau foetal (milieu CHO SFM II, Gibco). Le surnageant est ajusté à pH 8 et paessé sur une colonne de DEAE-sépharose (Pharmacia). La colonne est lavée avec un tampon 50 mM Tris, pH 8, 0,2 M NaCI. La molécule ESM-1 est éluée dans un tampon 50 mM Tris, pH 8, 1 M NaCI. L'éluat est ensuite dilué au 1 :4 dans un tampon 50 mM Tris, pH 8 et incubé en présence d'anticorps monoclonal anti-ESM-1 (MEC4) immobilisé sur agarose (Biorad). Après une nuit d'incubation à 4°C sous agitation les billes d'agarose sont lavées avec le tampon 50 mM Tris, pH 8, 0,2 M NaCI. ESM-1 est élue avec 3 M MgCI2. L'éluat est concentré et dialyse dans le tampon 50 mM Tris, pH 8, 0.5 M NaCI et stocké à -70°C. Des souris Balb/C ont été immunisées par injection de 10 μm de protéine ESM-1 recombinante purifiée par souris, selon un protocole d'immunisation standard en présence d'adjuvant de Freund.The ESM-1 cDNA is inserted into the eukaryotic expression vector pcDNA3 (In vitrogen) and then transfected into CHO cells with lipofectamine (GIBCO) according to the manufacturer's recommendations. 48 hours after transfection, the cells are subcultured in the presence of a selection agent (G418, Gibco) at a dose of 1000 micrograms / ml). After two weeks of selection, the G418-resistant CHO cells are given by limiting dilution. Clones expressing ESM-1 are then selected and called CHO-ESM (deposited at the CNCM). For production, CHO-ESM cells are cultured in suspension in a conditioned medium without fetal calf serum (CHO SFM II medium, Gibco). The supernatant is adjusted to pH 8 and weighed on a DEAE-sepharose column (Pharmacia). The column is washed with a 50 mM Tris buffer, pH 8, 0.2 M NaCl. The ESM-1 molecule is eluted in a 50 mM Tris buffer, pH 8, 1 M NaCl. The eluate is then diluted 1: 4 in a 50 mM Tris buffer, pH 8 and incubated in the presence of anti-ESM-1 monoclonal antibody (MEC4) immobilized on agarose (Biorad). After an overnight incubation at 4 ° C. with agitation, the agarose beads are washed with the 50 mM Tris buffer, pH 8, 0.2 M NaCl. ESM-1 is elected with 3M MgCI 2 . The eluate is concentrated and dialyzed in the 50 mM Tris buffer, pH 8, 0.5 M NaCl and stored at -70 ° C. Balb / C mice were immunized by injection of 10 μm of recombinant protein ESM-1 purified by mouse, according to a standard immunization protocol in the presence of Freund's adjuvant.
Les cellules d'hybridome sécrétant les anticorps monoclonaux anti-ESM-1 ont été obtenues par fusion, criblage et sous-clonage selon la technique décrite par BECHARD et al. (2000).Hybridoma cells secreting anti-ESM-1 monoclonal antibodies were obtained by fusion, screening and subcloning according to the technique described by BECHARD et al. (2000).
Cinq clones cellulaires d'hybridome ont été obtenus et ont été désignés génériquement MEC (« Mouse Monoclonal Antibody to ESM-1 produced by CHO Cells »). Quatre des hybridomes sélectionnés sont des isotypes lgG1 ,k respectivement les hybridomes désignés MEC4, MEC5, MEC15 et MEC36.Five cell clones of hybridoma were obtained and were generically designated MEC ("Mouse Monoclonal Antibody to ESM-1 produced by CHO Cells"). Four of the hybridomas selected are isotypes lgG1, k respectively the hybridomas designated MEC4, MEC5, MEC15 and MEC36.
L'un des hybridomes est l'isotype IgM.k, l'hybridome MEC11.One of the hybridomas is the IgM.k isotype, the MEC11 hybridoma.
Les clones cellulaires d'hybridomes ont été cultivés dans du milieu de culture en l'absence de sérum et les anticorps anti-ESM-1 ont été purifiés par chromatographie sur colonne de protéine G-Sépharose commercialisée par la Société Pharmacia (UPSALA, Suède). The cell clones of hybridomas were cultured in culture medium in the absence of serum and the anti-ESM-1 antibodies were purified by column chromatography on protein G-Sepharose sold by the company Pharmacia (UPSALA, Sweden) .
EXEMPLE 4:EXAMPLE 4:
Préparation d'un composé antagoniste de la protéine ESM-1 du type polypeptidePreparation of an antagonist compound of the ESM-1 protein of the polypeptide type
La mutagenèse dirigée a été réalisée avec le kit commercialisé par la Société STRATAGENE sous la référence « Site-directed quick mutagenesis kit », qui a été utilisé selon les recommandations du fabricant.Directed mutagenesis was carried out with the kit marketed by the company STRATAGENE under the reference "Site-directed quick mutagenesis kit", which was used according to the manufacturer's recommendations.
Brièvement, un couple d'amorces aller et retour de séquences strictement complémentaire sont synthétisées, ces amorces comprenant les nucléotides codant pour le ou les acides aminés mutés, ou les nucléotides complémentaires, ces nucléotides étant localisés au centre de la séquence des amorces qui comprennent aussi environ 10 à 15 nucléotides consécutifs complémentaires de la séquence à amplifier à la fois en 5' et en 3' des nucléotides centraux.Briefly, a pair of round-trip primers of strictly complementary sequences are synthesized, these primers comprising the nucleotides coding for the mutated amino acid (s), or the complementary nucleotides, these nucleotides being located at the center of the sequence of the primers which also include approximately 10 to 15 consecutive nucleotides complementary to the sequence to be amplified both 5 'and 3' of the central nucleotides.
Après amplification par PCR, les polynucléotides amplifiés codant pour la protéine ESM-1 mutée sont insérés dans le vecteur pCDNA3.After amplification by PCR, the amplified polynucleotides encoding the mutated ESM-1 protein are inserted into the vector pCDNA3.
On a utilisé respectivement les couples d'amorces suivants:The following pairs of primers were used respectively:
a) Pour la protéine ESM-1 F115Aa) For the protein ESM-1 F115A
Amorce aller : 5'-GCC TGA AAT TCC CCG CCT TCC AAT ATT CAG-3' ( SEQ ID N°3).Primer go: 5'-GCC TGA AAT TCC CCG CCT TCC AAT ATT CAG-3 '(SEQ ID N ° 3).
Amorce retour: 5'-CTG AAT ATT GGA AGG CGG GGA ATT TCA GGC- 3'Return bait: 5'-CTG AAT ATT GGA AGG CGG GGA ATT TCA GGC- 3 '
( SEQ ID N°4).(SEQ ID N ° 4).
b) Pour la protéine ESM-1 F116Ab) For the protein ESM-1 F116A
Amorce aller : 5'-CCT GAA ATT CCC CTT CGC CCA ATA TTC AGT AAC C-3' (SEQ ID N°5). Amorce retour: 5'-GGT TAC TGA ATA TTG CGC GAA GGG GAA TTT CAGT G-3' (SEQ ID N°6).Primer go: 5'-CCT GAA ATT CCC CTT CGC CCA ATA TTC AGT AAC C-3 '(SEQ ID N ° 5). Return primer: 5'-GGT TAC TGA ATA TTG CGC GAA GGG GAA TTT CAGT G-3 '(SEQ ID N ° 6).
c) Pour la protéine ESM-1 F115 F116Ac) For the protein ESM-1 F115 F116A
Amorce aller: 5'- CCT GAA ATT CCC CGC CGC CCA ATA TTC AGT AAC C-3' (SEQ ID N°7).Bait go: 5'- CCT GAA ATT CCC CGC CGC CCA ATA TTC AGT AAC C-3 '(SEQ ID N ° 7).
Amorce retour: 5'- GGT TAC TGA ATA TTG GGC GGC GGG GAA TTT CAG G-3'- (SEQ ID N°8).Return primer: 5'- GGT TAC TGA ATA TTG GGC GGC GGG GAA TTT CAG G-3'- (SEQ ID N ° 8).
EXEMPLE 5:Activité pro-tumoriαène de la protéine ESM-1 glycosylée.EXAMPLE 5 Pro-tumoriαene activity of the glycosylated protein ESM-1.
A. MATERIELS et METHODESA. MATERIALS AND METHODS
A.1. Lignées cellulaires:HEK T. HEK ESM/WT, HEK ESM/S137A, HEK ESM/69, HEK ESM/71 , HEK ESM/73.A.1. Cell lines: HEK T. HEK ESM / WT, HEK ESM / S137A, HEK ESM / 69, HEK ESM / 71, HEK ESM / 73.
On a utilisé la lignée cellulaire nommée HEK ESM/WT transfectée de façon stable avec l'ADNc codant pour la forme sauvage de ESM-1 (ESM/WT). Quatre autres lignées cellulaires sont obtenues par transfection avec de l'ADNc codant pour des formes purifiées de ESM-1 obtenues par mutagenèse dirigée à partir de la forme sauvage. La première d'entre elle, appelée HEK ESM/S137A, exprime la protéine ESM-1 mutée, non glycosylée, où une alanine est substituée à la serine 137, site majeur de O-glycosylation. Les trois autres lignées exprimant une forme glycosylée de ESM-1 dont la partie protéique est mutée. Il s'agit des lignées HEK ESM/F1 15A (remplacement de la phénylalanine en position 134, HEK ESM/71 (remplacement de la phénylalanine en position 135) et HEK ESM/F1 15A, F1 16A (double délétion remplacement 134-135).The cell line named HEK ESM / WT was transfected stably with the cDNA encoding the wild form of ESM-1 (ESM / WT). Four other cell lines are obtained by transfection with cDNA encoding purified forms of ESM-1 obtained by site-directed mutagenesis from the wild form. The first of these, called HEK ESM / S137A, expresses the mutated, non-glycosylated protein ESM-1, where an alanine is substituted for serine 137, the major site of O-glycosylation. The other three lines expressing a glycosylated form of ESM-1 whose protein part is mutated. These are the lines HEK ESM / F1 15A (replacement of phenylalanine in position 134, HEK ESM / 71 (replacement of phenylalanine in position 135) and HEK ESM / F1 15A, F1 16A (double deletion replacement 134-135) .
Ainsi, six lignées cellules produisant différentes formes de ESM- 1 sont utilisées: - HEK témoin, ne sécrétant pas de ESM-1 ; - Forme sauvage de ESM-1: HEK ESM/WT;Thus, six cell lines producing different forms of ESM-1 are used: - control HEK, not secreting ESM-1; - Wild form of ESM-1: HEK ESM / WT;
- Forme déglycosylée de ESM-1: HEK ESM/S137A;- Deglycosylated form of ESM-1: HEK ESM / S137A;
- Formes glycosylées dont la partie protéique est mutée au niveau de la région 115-116; HEK-ESM/69, HEK-ESM/71 , HEK ESM/73.- Glycosylated forms of which the protein part is mutated at the level of the region 115-116; HEK-ESM / 69, HEK-ESM / 71, HEK ESM / 73.
A2. Modèle murin de tumeurs xénogéniquesA2. Mouse model of xenogenic tumors
Les souris utilisées sont de type SCID (Severe Combined IMMUNO Déficience). Il s'agit plus précisément de souris C.B.17 Scid/scid fournies par l'animalerie de l'Institut Pasteur de Lille. Ces souris présentent une mutation autosomale récessive au niveau de leur système de recombinaison (Blunt., 1995). Cette mutation a pour conséquence la production d'immunoglobulines et de récepteurs de cellules T (TcR) et B 5BcR) non fonctionnels. De ce fait, elles ne possèdent pas de lymphocytes T et B fonctionnels; ces souris tolèrent donc le non-soi et représentent un modèje . de choix pour le développement de tumeurs xénogéniques. Les souris SCID utilisées sont des souris mâles et jeunes puisque âgées de 3 à 5 semaines. Pour chacune d'entre-elles, une injection intra-péritonéale d'anticorps anti- ascialo GM-1 (100 μg par souris dilué dans 200 μl de RPMI) est réalisée 24 heures avant l'injection des différentes lignées cellulaires. Il s'agit d'anticorps polyclonaux de lapin (Wako Pure Chemical Industries, Ltd) dirigés spécifiquement contre l'antigène asialo GM-1 exprimé par les cellules NK. Des travaux ont montré que l'utilisation de ces anticorps dans des modèles murins permet de neutraliser l'effet cytotoxique de cellules NK et de favoriser la greffe tumorale (Mather G et al. (1994).The mice used are of the SCID (Severe Combined IMMUNO Deficiency) type. More specifically, they are C.B.17 Scid / scid mice supplied by the animal facility of the Institut Pasteur in Lille. These mice have an autosomal recessive mutation in their recombination system (Blunt., 1995). This mutation results in the production of immunoglobulins and non-functional T cell receptors (TcR) and B 5BcR). As a result, they do not have functional T and B lymphocytes; these mice therefore tolerate non-self and represent a model. of choice for the development of xenogenic tumors. The SCID mice used are male and young mice since they are 3 to 5 weeks old. For each of them, an intraperitoneal injection of anti-ascialo GM-1 antibodies (100 μg per mouse diluted in 200 μl of RPMI) is carried out 24 hours before the injection of the various cell lines. These are rabbit polyclonal antibodies (Wako Pure Chemical Industries, Ltd) directed specifically against the asialo GM-1 antigen expressed by NK cells. Studies have shown that the use of these antibodies in mouse models makes it possible to neutralize the cytotoxic effect of NK cells and to promote tumor grafting (Mather G et al. (1994).
Quatre lots de souris (10 à 15 souris par groupe) anesthésiées à l'éther, sont ensuite injectés, par voie sous-cutanée au niveau du dos. Chaque souris reçoit 1 million de cellules diluées dans 200 μl de RPMI. L'injection de ces cellules définit le premier jour de l'expérience (Jo). Pour chaque souris, une surveillance macroscopique du point d'injection à la recherche de l'apparition d'une éventuelle tumeur ainsi qu'une mesure du poids corporel sont réalisées de façon hebdomadaire. A partir de la 5ème semaine, les souris sont saignées (environ 500 μl par souris), une fois par semaine, afin de doser les taux sériques de ESM-1 par un test ELISA (BECHARD D et al., 2000). Pour chaque souris, un examen anatomo-pathologique est réalisé.Four batches of mice (10 to 15 mice per group) anesthetized with ether are then injected, subcutaneously at the back. Each mouse receives 1 million cells diluted in 200 μl of RPMI. The injection of these cells defines the first day of the experiment (Jo). For each mouse, macroscopic monitoring of the injection site in search of the appearance of a possible tumor as well as a measurement of the body weight are carried out weekly. From the 5th week, the mice are bled (approximately 500 μl per mouse), once a week, in order to measure the serum levels of ESM-1 by a ELISA test (BECHARD D et al., 2000). For each mouse, an anatomo-pathological examination is carried out.
B. RESULTATSB. RESULTS
B.1.Induction de tumeurs chez la souris par la protéine ESM-1 glycosylée.B.1. Induction of tumors in mice by the glycosylated protein ESM-1.
Des cellules HEK ont été transfectees par un vecteur possédant un insert contenant l'ADNc codant pour la protéine ESM-1 sauvage glycosylée, désignée ESM/WT. Les cellules HEK sont injectées par voie sous-cutanée à des souris SCID âgées de 5 semaines. Chaque souris reçoit . préalablement une injection d'anticorps anti-asialo GN-1 par voie intrapéritonéale. On a analysé le pourcentage de tumeurs ayant un volume supérieur à 1 cm3 observé chez les souris à la huitième semaine suivant l'injection des cellules HEK transfectees.HEK cells were transfected with a vector having an insert containing the cDNA coding for the glycosylated wild-type protein ESM-1, designated ESM / WT. HEK cells are injected subcutaneously into 5 week old SCID mice. Each mouse receives. previously injected anti-asialo GN-1 antibody intraperitoneally. The percentage of tumors with a volume greater than 1 cm 3 observed in the mice at the eighth week following the injection of the transfected HEK cells was analyzed.
Les résultats sont représentés sur la figure 7.The results are shown in Figure 7.
A la figure 7A, on observe que l'injection de cellules HEK témoins n'induit pas l'apparition de tumeurs chez la souris. En revanche, les cellules HEK transfectees avec un ADN codant pour la protéine ESM- 1 glycosylée induisent de nombreuses tumeurs macroscopiquement visibles, dont approximativement 95% d'entre elles ont un volume tumoral supérieur à 1 cm3. La figure 7B illustre la cinétique d'apparition des tumeurs chez des souris qui ont reçu les cellules HEK transfectees avec un ADN codant pour la protéine ESM-1 glycosylée. On peut observer que le volume tumoral moyen, exprimé en cm3, croît continuellement dès la quatrième semaine suivant l'injection des cellules HEK transfectees. Les résultats expérimentaux présentés à la figure 7 démontrent clairement que la protéine ESM-1 glycosylée a une activité pro-tumorale.In FIG. 7A, it is observed that the injection of HEK control cells does not induce the appearance of tumors in the mouse. In contrast, HEK cells transfected with DNA encoding the glycosylated protein ESM-1 induce many macroscopically visible tumors, of which approximately 95% of them have a tumor volume greater than 1 cm 3 . FIG. 7B illustrates the kinetics of appearance of tumors in mice which have received HEK cells transfected with DNA coding for the glycosylated protein ESM-1. It can be observed that the average tumor volume, expressed in cm 3 , increases continuously from the fourth week following the injection of the transfected HEK cells. The experimental results presented in FIG. 7 clearly demonstrate that the glycosylated protein ESM-1 has a pro-tumor activity.
On a également mesuré les taux sériques de protéine ESM-1 chez des souris ayant reçu les cellules HEK témoins et chez les souris ayant reçu les cellules HEK transfectees par l'ADNc codant pour la protéine ESM-1. Les résultats sont représentés sur la figure 8.ESM-1 protein serum levels were also measured in mice having received control HEK cells and in mice having received HEK cells transfected with the cDNA encoding protein ESM-1. The results are shown in Figure 8.
Les résultats illustrés à la figure 8A montrent que la protéine ESM-1 n'est pas retrouvée dans les sérums des souris ayant reçu les cellules HEK témoins. Au contraire, un taux sérique de 40 à 50 nanogrammes par ml est retrouvé chez les souris ayant reçu les cellules HEK transfectees avec l'ADNc codant pour la protéine ESM-1 à la huitième semaine suivant l'injection des cellules.The results illustrated in FIG. 8A show that the ESM-1 protein is not found in the sera of the mice having received the HEK control cells. On the contrary, a serum level of 40 to 50 nanograms per ml is found in mice having received HEK cells transfected with the cDNA coding for the protein ESM-1 at the eighth week following the injection of the cells.
On a aussi analysé la cinétique des taux sériques de ESM-1 chez les souris ayant reçu la cellule HEK transfectée exprimant la protéine ESM-1 glycosylée (ESM/WT).The kinetics of the serum ESM-1 levels were also analyzed in the mice having received the transfected HEK cell expressing the glycosylated protein ESM-1 (ESM / WT).
Les résultats sont représentés à la figure 8B.The results are shown in Figure 8B.
On peut observer qu'une quantité détectable de protéine ESM-1 est retrouvée dans le sérum des souris dès la cinquième semaine suivant l'injection des cellules et que le taux sérique s'accroît rapidement et continuellement de la cinquième à la douzième semaine suivant l'injection des cellules.It can be observed that a detectable amount of protein ESM-1 is found in the serum of the mice from the fifth week following the injection of the cells and that the serum level increases rapidly and continuously from the fifth to the twelfth week following the injection of cells.
Les résultats expérimentaux illustrés à la figure 8 montrent que les tumeurs qui se sont développées chez les souris ayant reçu les cellules HEK transfectees produisent la protéine ESM-1. De plus, la quantité de protéine ESM-1 produite dans la circulation suit la cinétique de développement des tumeurs chez les souris.The experimental results illustrated in FIG. 8 show that the tumors which have grown in mice having received the transfected HEK cells produce the protein ESM-1. In addition, the amount of ESM-1 protein produced in the circulation follows the kinetics of tumor development in mice.
EXEMPLE 6:EXAMPLE 6
Activité pro-tumoriqène de différentes formes de la protéine ESM-1..Pro-tumor activity of different forms of the ESM-1 protein.
A. MATERIELS ET METHODESA. MATERIALS AND METHODS
Les matériels et méthodes utilisés dans cet exemple sont identiques à ceux décrits pour l'exemple 5.The materials and methods used in this example are identical to those described for example 5.
B. RESULTATSB. RESULTS
Les cellules HEK ont été transfectees respectivement par des vecteurs possédant un insert d'ADN codant respectivement pour la forme sauvage de ESM-1 (ESM/WT), une forme non glycosylée de ESM-1 (ESM/S137A) et une forme glycosylée de ESM-1 mutée au niveau des résidus phénylalanine en positions 134 et 135 qui ont été remplacés chacun par un résidu alanine (ESM/73). Les différentes cellules transfectees ont été injectées par voie sous-cutanée à des souris SCID âgées de 5 semaines et ayant préalablement reçues des anticorps anti- asialo GM-1.The HEK cells were transfected respectively by vectors having a DNA insert coding respectively for the wild form of ESM-1 (ESM / WT), a non-glycosylated form of ESM-1 (ESM / S137A) and a glycosylated form of ESM-1 mutated at the phenylalanine residues in positions 134 and 135 which have each been replaced by an alanine residue (ESM / 73). The different transfected cells were injected subcutaneously into 5-week-old SCID mice having previously received anti-asialo GM-1 antibodies.
On a analysé le pourcentage de tumeurs macroscopiquement visible ayant un volume tumorale supérieur à 1 cm3 chez les différents lots de souris. Les résultats sont représentés à la figure 9A. Les résultats de la figure 9A montrent que seule la protéineThe percentage of macroscopically visible tumors having a tumor volume greater than 1 cm 3 was analyzed in the different batches of mice. The results are shown in Figure 9A. The results of FIG. 9A show that only the protein
ESM-1 glycosylée est capable d'induire des tumeurs chez la souris. Ni la protéine ESM-1 non glycosylée, ni la protéine ESM-1 glycosylée mais mutée sur les résidus phénylalanine en positions 134 et 135 ne sont capables d'induire le développement de tumeurs chez les souris SCID. On a également analysé le taux sérique de protéine ESM-1 circulante dans les différents lots de souris. Les résultats sont représentés à la figure 9B.Glycosylated ESM-1 is capable of inducing tumors in mice. Neither the non-glycosylated ESM-1 protein nor the glycosylated but mutated ESM-1 protein on the phenylalanine residues at positions 134 and 135 are capable of inducing the development of tumors in SCID mice. The serum level of circulating ESM-1 protein was also analyzed in the different batches of mice. The results are shown in Figure 9B.
Les résultats de la figure 9B montrent que des taux détectables de protéine ESM-1 sérique ne peuvent être mesurés, à la huitième semaine suivant l'injection des cellules, que chez les souris ayant reçu les cellules HEK exprimant la protéine ESM-1 glycosylée (ESM/WT).The results in FIG. 9B show that detectable levels of serum ESM-1 protein can be measured, at the eighth week following the injection of the cells, only in mice having received HEK cells expressing the glycosylated ESM-1 protein ( ESM / WT).
Ni les souris injectées avec des cellules exprimant la protéine ESM-1 non glycosylée (ESM/S137A), ni les souris ayant reçu les cellules HEK exprimant la protéine ESM-1 glycosylée et mutée HEK-ESM/F115A, F116A) ne produisent de protéine ESM-1.Neither the mice injected with cells expressing the non-glycosylated protein ESM-1 (ESM / S137A), nor the mice having received the HEK cells expressing the glycosylated and mutated protein ESM-1 HEK-ESM / F115A, F116A) do not produce protein ESM-1.
L'ensemble des résultats présentés dans cet exemple confirme le pouvoir pro-tumorigène de la protéine ESM-1 glycosylée.All the results presented in this example confirm the pro-tumorigenic power of the glycosylated protein ESM-1.
Les résultats montrent aussi que des formes non glycosylées de la protéine ESM-1 ou encore des formes mutées de la protéine ESM-1 peuvent se comporter comme des antagonistes de cette protéine et posséder un pouvoir préventif et/ou curatif à l'égard de pathologies cancéreuses. EXEMPLE 7:The results also show that non-glycosylated forms of the protein ESM-1 or even mutated forms of the protein ESM-1 can behave like antagonists of this protein and have a preventive and / or curative power with regard to pathologies. cancerous. EXAMPLE 7:
Dosage de la protéine ESM-1 circulante chez des patients atteints de cancers bronchopulmonaires à différents stades de développement. A. Matériels et méthode.Determination of the circulating ESM-1 protein in patients with bronchopulmonary cancers at different stages of development. A. Materials and method.
Le test d'immuno-détection consiste en un test immuno- enzymatique du type « sandwich » dont les caractéristiques générales sont identiques à celui décrit par BECHARD et al. (2000). L'anticorps monoclonal anti-ESM-1 produit par la lignée d'hybridomes MEP14 (CNCM N°l-1942) a été dilué à la concentration de 5μg/ml dans un tampon carbonate 0,1 M, pH 9,5, et adsorbé pendant une nuit à +4°C sur une plaque 96 puits (plaque E.I.A./R.IA, Costar, Cambridge, MA, USA). La plaque a été saturée pendant une heure à la température du laboratoire avec un volume de 200 μl/puits de tampon PBS contenant 0,1 % de sérum albumine bovine et 5mM de EDTA, puis lavée deux fois avec un tampon ELISA (le tampon PBS ci-dessus additionné de 0,1 % de Tween 20). Un calibrage a été réalisé avec la protéine ESM-1 purifiée selon la technique décrite par BECHARD et al. (2000).The immuno-detection test consists of an immuno-enzymatic test of the “sandwich” type, the general characteristics of which are identical to that described by BECHARD et al. (2000). The anti-ESM-1 monoclonal antibody produced by the MEP14 hybridoma line (CNCM No. l-1942) was diluted to a concentration of 5 μg / ml in 0.1 M carbonate buffer, pH 9.5, and adsorbed overnight at + 4 ° C on a 96-well plate (EIA / R.IA plate, Costar, Cambridge, MA, USA). The plate was saturated for one hour at laboratory temperature with a volume of 200 μl / well of PBS buffer containing 0.1% bovine serum albumin and 5 mM EDTA, then washed twice with ELISA buffer (PBS buffer above with 0.1% Tween 20 added). A calibration was carried out with the protein ESM-1 purified according to the technique described by BECHARD et al. (2000).
Les échantillons de sang ont été dilués en série (1 :2 à 1 :128), dans un tampon ELISA et incubés sur une plaque ELISA pendant une heure à la température du laboratoire. Les puits ont été lavés trois fois avec un tampon ELISA puis incubés pendant 1 heure à la température du laboratoire avec un second anticorps monoclonal dirigé contre ESM-1 , l'anticorps MEC15 (CNCM N°l-2572) à la concentration de 0,1 μg/ml dans 100 μl de tampon par puits. Après trois lavages, on a ajouté un anticorps monoclonal de rat biotinylé dirigé contre les lgG1 de souris (commercialisé par PHARMINGEN) dilué dans un tampon ELISA et laissé incuber ce second anticorps pendant une heure .The blood samples were diluted serially (1: 2 to 1: 128) in ELISA buffer and incubated on an ELISA plate for one hour at laboratory temperature. The wells were washed three times with ELISA buffer and then incubated for 1 hour at laboratory temperature with a second monoclonal antibody directed against ESM-1, the antibody MEC15 (CNCM No. 1-2572) at the concentration of 0, 1 μg / ml in 100 μl of buffer per well. After three washes, a biotinylated rat monoclonal antibody directed against mouse lgG1 was added (marketed by PHARMINGEN) diluted in ELISA buffer and allowed to incubate this second antibody for one hour.
Après trois lavages dans le tampon ELISA, les puits ont été incubés avec un conjugué de streptavidine-peroxydase à la dilution 1 :10.000 v/v (commercialisé par la Société ZYMED).After three washes in the ELISA buffer, the wells were incubated with a streptavidin-peroxidase conjugate at a dilution of 1: 10,000 v / v (sold by the company ZYMED).
Après 30 minutes d'incubation avec le conjugué streptavidine- peroxydase, on réalise trois lavages de chaque puits dans un tampon ELISA puis deux lavages dans un tampon PBS.After 30 minutes of incubation with the streptavidin-peroxidase conjugate, three washes of each well are carried out in an ELISA buffer and then two washes in a PBS buffer.
Le conjugué streptavidine-peroxydase est révélé avec le substrat TMB commercialisé par la Société SIGMA (Saint-Louis, MO, USA) en présence de 255 μl de H202 pendant 30'.The streptavidin-peroxidase conjugate is revealed with the TMB substrate sold by the SIGMA company (Saint-Louis, MO, USA) in the presence of 255 μl of H 2 0 2 for 30 '.
La réaction de révélation est stoppée par l'addition d'un volume de 100μl de H2SO4 2N.The revelation reaction is stopped by the addition of a volume of 100 μl of 2N H 2 SO 4 .
La plaque est lue à l'aide d'un spectrophotomètre (anthos labtec LP40, France) à la longueur d'onde de 405 nanomètres.The plate is read using a spectrophotometer (anthos labtec LP40, France) at the wavelength of 405 nanometers.
La concentration de la protéine ESM-1 plasmatique ou sérique est calculée à partir des mesures de densité optique et exprimée en nanogrammes par ml.The concentration of the plasma or serum ESM-1 protein is calculated from the optical density measurements and expressed in nanograms per ml.
B. RESULTATSB. RESULTS
On a mesuré la concentration de protéine ESM-1 circulante dans le sérum de différents patients porteurs d'un cancer bronchopulmonaire à différents stades de développement, respectivement au stade I, II, IIIA, IIIB et IV selon la classification internationale TNM qui est définie ci-après:The concentration of circulating ESM-1 protein in the serum of different patients with bronchopulmonary cancer was measured at different stages of development, respectively in stages I, II, IIIA, IIIB and IV according to the international TNM classification which is defined below. -after:
T = taille de la tumeur (T1 : < 1 cm; T2: entre 1 et 3 cm; T3:> 3 cm.T = tumor size (T1: <1 cm; T2: between 1 and 3 cm; T3:> 3 cm.
N = nodule ganglionnaire (NO si non envahi; N1 si envahi). M = métastase à distance (MO si pas de méastase; M si métastase). Les patients atteints d'un cancer au stade I présentent une concentration sérique de protéine ESM-1 de 1 ,43 +/- 0,76 nanogrammes/ml (n = 3).N = lymph node (NO if not invaded; N1 if invaded). M = distant metastasis (MO if no meastasis; M if metastasis). Patients with stage I cancer have a serum ESM-1 concentration of 1.43 +/- 0.76 nanograms / ml (n = 3).
Les patients atteints d'un cancer bronchopulmonaire au stade II présentent une concentration sérique de la protéine ESM-1 de 0,72 +/- 0,39 nanogrammes/ml (n = 3).Patients with stage II bronchopulmonary cancer have a serum ESM-1 concentration of 0.72 +/- 0.39 nanograms / ml (n = 3).
Les patients atteints d'un cancer bronchopulomonaire au stade Illa présentent une concentration de protéine ESM-1 circulante de 0,9 +/- 0,53 nanogrammes/ml (n= 2). Les patients atteints d'un cancer bronchopulmonaire au stadePatients with Illa stage bronchopulomonary cancer have a circulating ESM-1 protein concentration of 0.9 +/- 0.53 nanograms / ml (n = 2). Patients with stage bronchopulmonary cancer
IIIB présentent une concentration de protéine ESM-1 circulante de 3,1 +/- 2,17 nanogrammes/ml (n = 3).IIIB have a circulating ESM-1 protein concentration of 3.1 +/- 2.17 nanograms / ml (n = 3).
Les patients atteints d'un cancer bronchopulmonaire au stade IV présentent une concentration de protéine ESM-1 circulante de 3,1 +/- 1 ,91 nanogrammesd/ml (n = 11 ).Patients with stage IV bronchopulmonary cancer have a circulating ESM-1 protein concentration of 3.1 +/- 1.91 nanogramsd / ml (n = 11).
Les résultats présentés ci-dessus montrent que le taux sérique de protéine ESM-1 augmente en fonction du stade d'évolution du cancer.The results presented above show that the serum ESM-1 protein level increases as the stage of cancer progresses.
Une relation claire est donc démontrée entre le niveau de production de la protéine ESM-1 dans la circulation sanguine et la sévérité d'un cancer chez un patient.A clear relationship is therefore demonstrated between the level of production of the protein ESM-1 in the blood circulation and the severity of cancer in a patient.
EXEMPLE 8 : Activité anti-tumorale d'un composé antagoniste de ESM-1 du type anticorps.EXAMPLE 8 Anti-tumor activity of an ESM-1 antagonist compound of the antibody type.
A. MATERIEL ET METHODE Les anticorps monoclonaux MEP-08 sont injectés par voie intrapéritonéale à la dose de 400 μg à partir de la deuxième suivant l'inoculation des cellules HEK ESM-WT. Les injections sont répétées de façon hebdomadaire pendant 12 semaines. Un anticorps contrôle, MEP- 14, est utilisé dans les mêmes conditions. Les souris sont sacrifiées lorsque leur volume tumoral est supérieur à 6 cm3, (n > 8 souris dans chaque groupe). La figure représente le pourcentage de souris vivantes dans chacun des groupes.A. MATERIAL AND METHOD The monoclonal antibodies MEP-08 are injected intraperitoneally at a dose of 400 μg from the second following inoculation of the HEK ESM-WT cells. The injections are repeated weekly for 12 weeks. A control antibody, MEP-14, is used under the same conditions. The mice are sacrificed when their tumor volume is greater than 6 cm 3 , (n> 8 mice in each group). The figure represents the percentage of live mice in each of the groups.
B. RESULTATS Dans la mesure où la phénylalanine en position 1 15 est indispensable au développement tumoral, elle constitue une nouvelle cible thérapeutique. C'est la raison pour laquelle les anticorps monoclonaux MEP-08 anti-ESM-1 , produits par la lignée d'hybridome MEP-08 déposée à la CNCM sous le n°l-1941 , dirigés spécifiquement contre cette région ont été produits et injectés au groupe de souris HEK- ESM/WT. Ils ont pour but d'étudier le rôle du peptide de ESM-1 dans le développement tumoral et d'évaluer un éventuel effet thérapeutique. Afin d'éliminer un effet anti-tumoral dépendant du fragment Fc de l'anticorps (réaction d'ADCC), un anticorps contrôle de même isotype mais reconnaissant un épitope différent a été utilisé parallèlement et dans les mêmes conditions.B. RESULTS Insofar as phenylalanine in position 1 15 is essential for tumor development, it constitutes a new therapeutic target. This is the reason why the anti-ESM-1 monoclonal antibodies MEP-08, produced by the hybridoma line MEP-08 deposited at the CNCM under the number l-1941, directed specifically against this region were produced and injected into the group of HEK-ESM / WT mice. Their aim is to study the role of the ESM-1 peptide in tumor development and to evaluate a possible therapeutic effect. In order to eliminate an anti-tumor effect dependent on the Fc fragment of the antibody (ADCC reaction), a control antibody of the same isotype but recognizing a different epitope was used in parallel and under the same conditions.
La figure 10 montre que les injections précoces d'anticorps MEP-08 augmentent la survie des souris de façon significative de près de 60% alors que les anticorps MEP-14 n'ont pas d'effet. Ces premiers résultas montrent qu'il s'agit d'une action spécifique liée au fragment Fab de l'anticorps dirigé spécifiquement contre la phénylalanine en position 115 et confirment l'implication du peptide dans la croissance tumorale. Il est surprenant de constater que cet effet sur la survie diminue lorsque les anticorps sont administrés de façon retardée. Quelle que soit la semaine où débutent les injections, les anticorps peuvent retarder ou empêcher la croissance tumorale. Cet effet anti-tumoral reste plus prononcé lorsque les anticorps sont utilisés précocement. REFERENCES BIBLIOGRAPHIQUES:Figure 10 shows that early injections of MEP-08 antibodies significantly increase mouse survival by almost 60% while MEP-14 antibodies have no effect. These first results show that it is a specific action linked to the Fab fragment of the antibody directed specifically against phenylalanine at position 115 and confirm the involvement of the peptide in tumor growth. It is surprising to find that this effect on survival decreases when antibodies are administered in a delayed manner. No matter which week the injections start, antibodies can delay or prevent tumor growth. This anti-tumor effect remains more pronounced when the antibodies are used early. BIBLIOGRAPHICAL REFERENCES:
ANFOSSI et al. (1989), Acad. Sci., vol.86: 3379-3383 AUSUBEL F. et al.K, 1989, Current Protocols in Molecular Biology GREEN publishing Associates and WHILEY Interscience, N.Y. BECHARD et al. 2000, J. Vase. Res. 37(5): 417-425. BEAUCAGE et al., 1981, Tetrahedron Lett., vol.22: 1859-1862 BLUNT (1995) [Référence à compléter S.V.P.]. BROWN EL, BELAGAJE R, RYAN MJ, KHORANA HG, Methods ENZYMOL, 1979, Vol. 68: 109-151 BUSH et al., 1997, J. Chromatogr. , vol.777: 311-328 CELLA G et al., 1992, Angiology, vol.43 DELORME MA et al., (1996, Thromb Haemost, vol.75:747-751.) EDWARDS and LEATHERBARROW, 1997, Analytical Biochemistry, vol. 246: 1 - 6 . FELICI F, 1991, J. Mol. Biol., vol. 222:301-310 GAO et al, 1996, J. Biol. Chem, vol. 271 (15):9002-9008 GLUZMAN , 1982, Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory.ANFOSSI et al. (1989), Acad. Sci., Vol.86: 3379-3383 AUSUBEL F. et al. K, 1989, Current Protocols in Molecular Biology GREEN publishing Associates and WHILEY Interscience, N.Y. BECHARD et al. 2000, J. Vase. Res. 37 (5): 417-425. BEAUCAGE et al., 1981, Tetrahedron Lett., Vol.22: 1859-1862 BLUNT (1995) [Reference to be completed S.V.P.]. BROWN EL, BELAGAJE R, RYAN MJ, KHORANA HG, Methods ENZYMOL, 1979, Vol. 68: 109-151 BUSH et al., 1997, J. Chromatogr. , vol. 777: 311-328 CELLA G et al., 1992, Angiology, vol. 43 DELORME MA et al., (1996, Thromb Haemost, vol. 75: 747-751.) EDWARDS and LEATHERBARROW, 1997, Analytical Biochemistry , flight. 246: 1 - 6. FELICI F, 1991, J. Mol. Biol., Vol. 222: 301-310 GAO et al, 1996, J. Biol. Chem, vol. 271 (15): 9002-9008 GLUZMAN, 1982, Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory.
GREEN et al., 1986, Ann. Rev. Biochem., vol.55: 569-597 HOLT et al, 1988, Mol. Cell Biol. vol.8 : 963-973 HOUBEN WEYL, 1974, in Meuthode der Organischen Chemie, E.WUNSCH ed, vols.15-1 et 15-2GREEN et al., 1986, Ann. Rev. Biochem., Vol. 55: 569-597 HOLT et al, 1988, Mol. Cell Biol. vol.8: 963-973 HOUBEN WEYL, 1974, in Meuthode der Organischen Chemie, E.WUNSCH ed, vols.15-1 and 15-2
IZANT JG, WEINTRAUB H, 1984, Cell, vol.36 (4): 1007-1015 KOHLER G. and MIELSTEIN C. , 1975, Nat. Vol.256:495. KOZBOR et al. (1983), Hybridoma, vol.2 (1): 7-16 LASSALLE P et al. , 1996, The Journal of Biological Chemistry, vol.271(34): 20.458-20.464.IZANT JG, WEINTRAUB H, 1984, Cell, vol. 36 (4): 1007-1015 KOHLER G. and MIELSTEIN C., 1975, Nat. Vol.256: 495. KOZBOR et al. (1983), Hybridoma, vol. 2 (1): 7-16 LASSALLE P et al. , 1996, The Journal of Biological Chemistry, vol. 271 (34): 20.458-20.464.
LASSALLE P et al., 1992, Eur. J. Immunol., vol.22:425-431 LEGER et al. (1997), Hum. antibodies, vol.8 (1): 3-16. LUCAS AH, 1994, In: Development and Clinical uses of haemophilus b Conjuguate • MATHER G et al. 1994, Immunobiology, 190 (4-5): 333-345. MARTINEAU P, JONES P., WINTER G et al., 1998, J. Mol. Biol, vol.280(1): 117-127.LASSALLE P et al., 1992, Eur. J. Immunol., Vol. 22: 425-431 LEGER et al. (1997), Hum. antibodies, vol.8 (1): 3-16. LUCAS AH, 1994, In: Development and Clinical uses of haemophilus b Conjuguate • MATHER G et al. 1994, Immunobiology, 190 (4-5): 333-345. MARTINEAU P, JONES P., WINTER G et al., 1998, J. Mol. Biol, vol. 280 (1): 117-127.
MERRIFIELD RB, 1965a, Nat., vol.207(996): 522-523MERRIFIELD RB, 1965a, Nat., Vol. 207 (996): 522-523
MERRIFIELD RB 1965b Science, vol.150(693:178-185)MERRIFIELD RB 1965b Science, vol. 150 (693: 178-185)
MOXHAM et al., 1993, Science, vol.2602: 991-995.MOXHAM et al., 1993, Science, vol. 2602: 991-995.
NARANG SA, HSIUNG HN, BROUSSEAU R, et al. 1979, Methods enzymol, vol.68 ,90-98NARANG SA, HSIUNG HN, BROUSSEAU R, et al. 1979, Methods enzymol, vol. 68, 90-98
NATALI PG et al. , 1996, Int. J. Cancer, vol.69:212-217NATALI PG et al. , 1996, Int. J. Cancer, vol. 69: 212-217
LEE and NATHANS, 1988, J. Biol. Chem., vol.263: 3521LEE and NATHANS, 1988, J. Biol. Chem., Vol. 263: 3521
OLDENBURG KR et al., 1992, Proc Natl. Acad. Sci, vol. 89: 5393-OLDENBURG KR et al., 1992, Proc Natl. Acad. Sci, vol. 89: 5393-
53975397
RAHIMI N et al. (1998), J. Biol. Chem., vol.273:33.714-33.721RAHIMI N et al. (1998), J. Biol. Chem., Vol. 273: 33.714-33.721
RIDDER R, SCHMITZ R, LEGAY F, GRAM H, 1995, BiotechnologyRIDDER R, SCHMITZ R, LEGAY F, GRAM H, 1995, Biotechnology
(N.Y.), vol.13 (3): 255-260.(N.Y.), vol.13 (3): 255-260.
SIEGFRIED JM et al., 1998, Ann. Thorac. Surg.,vol.66: 1915-1918SIEGFRIED JM et al., 1998, Ann. Thorac. Surg., Vol. 66: 1915-1918
OTSUKA T et al., 1998, Cancer Res., vol.58:5157-5167OTSUKA T et al., 1998, Cancer Res., Vol. 58: 5157-5167
PARMLEY and SMITH, 1988, Gène, vol.73: 305-318PARMLEY and SMITH, 1988, Gene, vol. 73: 305-318
REINMANN KA et al., 1997, AIDS Res. Hum. Retroviruses, vol.13REINMANN KA et al., 1997, AIDS Res. Hmm. Retroviruses, vol.13
(11):933-943(11): 933-943
ROSSI et al., 1991, Pharmacol. Ther, vol.50 : 245-254ROSSI et al., 1991, Pharmacol. Ther, vol.50: 245-254
SALE et al, 1995, EMBO J. vol.14 (4): 674-684SALE et al, 1995, EMBO J. vol.14 (4): 674-684
SCZAKIEL G et al. , 1995, Trends Microbiol, vol.3 (6): 213-217SCZAKIEL G et al. , 1995, Trends Microbiol, vol.3 (6): 213-217
TAKAYAMA H ET Al, 1997, Lab. Invest. vol.77: 131-138TAKAYAMA H ET Al, 1997, Lab. Invest. vol. 77: 131-138
VALADON P et al. , 1996, J. Mol. Biol., vol. 261 : 11-22VALADON P et al. , 1996, J. Mol. Biol., Vol. 261: 11-22
WANG et al., 1997, Chromatographia, vol.44 : 205-208WANG et al., 1997, Chromatographia, vol. 44: 205-208
WESTERING MAJ, 1995, Proc. Natl. acad. sci, vol.92: 4021-4025WESTERING MAJ, 1995, Proc. Natl. acad. sci, vol. 92: 4021-4025
WICKSTROM et al, 1988, Proc. Natl. Acad. sci. vol.85: 1028-1032WICKSTROM et al, 1988, Proc. Natl. Acad. sci. vol. 85: 1028-1032
WEIDNER KM et al., 1993, J. Cell Biol. vol. 121 : 145-154 WEIDNER KM et al., 1993, J. Cell Biol. flight. 121: 145-154

Claims

REVENDICATIONS
1. Utilisation d'un composé antagoniste de la protéine ESM-1 pour la fabrication d'un médicament pour le traitement d'un cancer. 1. Use of a protein antagonist compound ESM-1 for the manufacture of a medicament for the treatment of cancer.
2. Utilisation selon la revendication 1 , caractérisée en ce que le composé antagoniste de la protéine ESM-1 consiste en un anticorps se liant spécifiquement à la protéine ESM-1.2. Use according to claim 1, characterized in that the antagonist compound of the protein ESM-1 consists of an antibody specifically binding to the protein ESM-1.
3. Utilisation selon la revendication 2, caractérisée en ce que le composé antagoniste du type anticorps se lie spécifiquement à l'un des déterminants antigéniques AgD1 , AgD2 ou AgD3 de la protéine ESM-1.3. Use according to claim 2, characterized in that the antagonist compound of the antibody type specifically binds to one of the antigenic determinants AgD1, AgD2 or AgD3 of the protein ESM-1.
4. Utilisation selon l'une des revendications 2 ou 3, caractérisée en ce que le composé antagoniste du type anticorps est choisi parmi les anticorps monoclonaux sécrétés par les lignées d'hybridome suivantes:4. Use according to one of claims 2 or 3, characterized in that the antagonist compound of the antibody type is chosen from monoclonal antibodies secreted by the following hybridoma lines:
- l'hybridome MEP21 déposé le 19 Novembre 1997 auprès de la Collection Nationale de Cultures des Micro-organismes de l'Institut- the MEP21 hybridoma deposited on November 19, 1997 with the National Collection of Cultures of Microorganisms of the Institute
Pasteur (CNCM) sous le n° d'accès 1-1944;Pasteur (CNCM) under access number 1-1944;
- l'hybridome MEP14 déposé le 19 Novembre 1997 auprès de la CNCM sous le n° d'accès n°l-1942;- the MEP14 hybridoma deposited on November 19, 1997 with the CNCM under the access number n ° l-1942;
- l'hybridome MEP19 déposé le 19 Novembre 1997 auprès de la CNCM sous le n° d'accès n°l-1943;- the MEP19 hybridoma deposited on November 19, 1997 with the CNCM under the access number n ° l-1943;
- l'hybridome MEP08 déposé le 19 Novembre 1997 auprès de la CNCM sous le N° d'accès n° 1-1941.- the MEP08 hybridoma deposited on November 19, 1997 with the CNCM under access number 1-1941.
5. Utilisation selon la revendication 2, caractérisée en ce que le composé antagoniste du type anticorps est l'anticorps produit par la lignée d'hybridome MEC15 déposée le 17 Octobre 2000 auprès de la CNCM sous le n° d'accès n°l-2572.5. Use according to claim 2, characterized in that the antagonist compound of the antibody type is the antibody produced by the hybridoma line MEC15 deposited on October 17, 2000 with the CNCM under the access number n ° l- 2572.
6. Utilisation selon la revendication 1 , caractérisée en ce que le composé antagoniste de la protéine ESM-1 est un polypeptide ayant une longueur d'au moins 10 acides aminés consécutifs de la séquence SEQ ID n°1 et comprenant la séquence d'acides aminés allant de l'acide aminé en position 119 jusqu'à l'acide aminé en position 139 de la séquence SEQ ID n°1 et comprenant au moins une substitution d'un acide aminé, par rapport à la séquence correspondante de la séquence SEQ ID N°1. 6. Use according to claim 1, characterized in that the antagonist compound of the protein ESM-1 is a polypeptide having a length of at least 10 consecutive amino acids of the sequence SEQ ID No. 1 and comprising the acid sequence amines ranging from amino acid in position 119 to amino acid in position 139 of the sequence SEQ ID No. 1 and comprising at least one substitution of an amino acid, relative to the corresponding sequence of the sequence SEQ ID # 1.
7. Utilisation selon la revendication 6, caractérisée en ce que le composé antagoniste de la protéine ESM-1 comprend une ou plusieurs substitutions d'un acide aminé contenant un cycle aromatique retrouvé dans la séquence SEQ ID n°1 par un acide aminé ne contenant pas de cycle aromatique.7. Use according to claim 6, characterized in that the antagonist compound of the protein ESM-1 comprises one or more substitutions of an amino acid containing an aromatic cycle found in the sequence SEQ ID n ° 1 by an amino acid not containing no aromatic cycle.
8. Utilisation selon l'une des revendications 6 ou 7, caractérisée en ce que le composé antagoniste de la protéine ESM-1 comprend, par rapport à la séquence SEQ ID N°1 , une substitution des résidus phénylalanine en positions 134 et 135 de la séquence SEQ ID N°1 par deux résidus d'acides aminés, identiques ou différents, ne contenant pas de cycle aromatique.8. Use according to one of claims 6 or 7, characterized in that the antagonist compound of the protein ESM-1 comprises, with respect to the sequence SEQ ID No. 1, a substitution of the phenylalanine residues in positions 134 and 135 of the sequence SEQ ID N ° 1 by two amino acid residues, identical or different, not containing an aromatic cycle.
9. Utilisation selon la revendication 1 , caractérisée en ce que le composé antagoniste de la protéine ESM-1 consiste en un oligonucléotide antisens. 9. Use according to claim 1, characterized in that the antagonist compound of the protein ESM-1 consists of an antisense oligonucleotide.
10. Utilisation selon la revendication 9, caractérisée en ce que le composé antagoniste de la protéine ESM-1 du type oligonucléotide antisens consiste en un polynucléotide comprenant au moins 20 nucléotides consécutifs de l'ADNc de ESM-1 de séquence SEQ ID N°2.10. Use according to claim 9, characterized in that the antagonist compound of the ESM-1 protein of the antisense oligonucleotide type consists of a polynucleotide comprising at least 20 consecutive nucleotides of the ESM-1 cDNA of sequence SEQ ID No. 2 .
11. Composé antagoniste de la protéine ESM-1 , caractérisé en ce qu'il consiste en l'anticorps monoclonal produit par la lignée d'hybridome MEC15 déposée auprès de la CNCM le 17 Octobre 2000 sous le n° d'accès n°l-2572.11. Compound antagonist of the protein ESM-1, characterized in that it consists of the monoclonal antibody produced by the hybridoma line MEC15 deposited with the CNCM on October 17, 2000 under the access number # l -2572.
12. Procédé de sélection d'un composé antagoniste de la protéine ESM-1 , caractérisé en ce qu'il comprend les étapes suivantes: a) injecter à un animal des cellules capables de former des tumeurs en présence de la protéine ESM-1 , lesdites cellules étant transfectees ou transformées par un acide nucléique capable d'exprimer la protéine ESM-1 in vivo; b) administrer à cet animal un composé candidat antagoniste de la protéine ESM-1 ; c) comparer la formation de tumeurs chez un premier animal tel qu'obtenu à la fin de l'étape b) et chez un second animal tel qu'obtenu à la fin de l'étape a); et d) sélectionner le composé candidat capable d'inhiber ou de bloquer la formation de tumeurs chez le premier animal. 12. A method of selecting an antagonist compound of the ESM-1 protein, characterized in that it comprises the following steps: a) injecting into an animal cells capable of forming tumors in the presence of the ESM-1 protein, said cells being transfected or transformed with a nucleic acid capable of expressing the protein ESM-1 in vivo; b) administering to this animal a candidate antagonist compound of the ESM-1 protein; c) comparing the formation of tumors in a first animal as obtained at the end of step b) and in a second animal as obtained at the end of step a); and d) selecting the candidate compound capable of inhibiting or blocking the formation of tumors in the first animal.
13. Procédé de sélection d'un composé candidat antagoniste de la protéine ESM-1 , caractérisé en ce qu'il comprend les étapes suivantes: a) fournir un polypeptide consistant en la protéine ESM-1 ou un fragment peptidique de cette protéine; b) mettre en contact ledit polypeptide avec le composé candidat à tester; c) détecter les complexes formés entre ledit polypeptide et le composé candidat; d) sélectionner les composés candidats se fixant sur le polypeptide constitué de la protéine ESM-1 ou d'un fragment de cette protéine.13. A method of selecting a candidate antagonist compound for the ESM-1 protein, characterized in that it comprises the following steps: a) providing a polypeptide consisting of the ESM-1 protein or a peptide fragment of this protein; b) bringing said polypeptide into contact with the candidate compound to be tested; c) detecting the complexes formed between said polypeptide and the candidate compound; d) selecting the candidate compounds which bind to the polypeptide consisting of the ESM-1 protein or of a fragment of this protein.
14. Procédé de sélection d'un composé antagoniste de la protéine ESM-1 , caractérisé en ce qu'il comprend les étapes suivantes: a) mettre en contact la protéine ESM-1 ou un fragment peptidique de cette dernière en présence de :14. Method for selecting an antagonist compound of the ESM-1 protein, characterized in that it comprises the following steps: a) bringing the ESM-1 protein or a peptide fragment of the latter into contact in the presence of:
(i) un composé antagoniste de la protéine ESM-1 se fixant sur la protéine ESM-1 ; et(i) an ESM-1 protein antagonist compound which binds to the ESM-1 protein; and
(ii) un composé candidat à tester; b) dans une étape séparée de l'étape a), mais éventuellement simultanée à cette dernière, mettre en contact la protéine ESM-1 ou un fragment peptidique de cette dernière avec un composé antagoniste de la protéine ESM-1 se fixant sur la protéine ESM-1 ; c) détecter la quantité respective du composé antagoniste de la protéine ESM-1 fixée à l'issue de chacune des étapes a) et b); et d) sélectionner le composé candidat qui entre en compétition avec le composé antagoniste pour la fixation sur la protéine ESM-1.(ii) a candidate compound to be tested; b) in a step separate from step a), but possibly simultaneous with the latter, bringing the ESM-1 protein or a peptide fragment of the latter into contact with an antagonist compound of the ESM-1 protein which binds to the protein ESM-1; c) detecting the respective amount of the antagonist compound of the ESM-1 protein fixed at the end of each of steps a) and b); and d) selecting the candidate compound which competes with the antagonist compound for binding to the ESM-1 protein.
15. Procédé pour sélectionner un composé antagoniste de la protéine ESM-1 à partir d'un composé candidat, caractérisé en ce qu'il comprend les étapes suivantes: a) sélectionner, parmi les composés candidats, les composés qui se fixent sur la protéine ESM-1 ou sur un fragment peptidique de cette protéine; b) administrer un composé sélectionné à l'étape a) à un animal et déterminer la capacité de ce composé à inhiber, chez cet animal, le développement de tumeurs induites par la protéine ESM-1 ; c) sélectionner les composés inhibiteurs du développement des tumeurs déterminés à l'étape b) comme des composés antagonistes de la protéine ESM-1.15. Method for selecting an antagonist compound of the ESM-1 protein from a candidate compound, characterized in that it comprises the following steps: a) selecting, from the candidate compounds, the compounds which bind to the protein ESM-1 or on a peptide fragment of this protein; b) administering a compound selected in step a) to an animal and determining the capacity of this compound to inhibit, in this animal, the development of tumors induced by the ESM-1 protein; c) selecting the compounds inhibiting the development of the tumors determined in step b) as antagonist compounds of the protein ESM-1.
16. Composition pharmaceutique pour le traitement et/ou la prévention d'un cancer comprenant un composé antagoniste de la protéine ESM-1. 16. Pharmaceutical composition for the treatment and / or prevention of cancer comprising a compound antagonist of the protein ESM-1.
17. Composition pharmaceutique selon la revendication 16, caractérisée en ce que le composé antagoniste de la protéine ESM-1 est un anticorps se liant spécifiquement à la protéine ESM-1.17. Pharmaceutical composition according to claim 16, characterized in that the ESM-1 protein antagonist compound is an antibody specifically binding to the ESM-1 protein.
18. Composition pharmaceutique selon la revendication 17, caractérisée en ce que le composé antagoniste de la protéine ESM-1 est l'anticorps produit par la lignée d'hybridome MEP08 déposée auprès de la CNCM le 19 Novembre 1997 sous le n° d'accès 1-1941 ou l'anticorps produit par la lignée d'hybridome MEC15 déposée auprès de la CNCM le 17 Octobre 2000 sous le N° d'accès I-2572.18. Pharmaceutical composition according to claim 17, characterized in that the antagonist compound of the protein ESM-1 is the antibody produced by the hybridoma line MEP08 deposited with the CNCM on November 19, 1997 under the access number 1-1941 or the antibody produced by the hybridoma line MEC15 deposited with the CNCM on October 17, 2000 under the access number I-2572.
19. Composition pharmaceutique selon la revendication 16, caractérisée en ce que le composé antagoniste de la protéine ESM-1 est un polypeptide comprenant au moins 10 acides aminés consécutifs de la protéine ESM-1 de séquence SEQ ID N°1 , comprenant une séquence d'acides aminés allant de l'acide aminé en position 119 jusqu'à l'acide aminé en position 139 de la séquence SEQ ID N°1 et comprenant au moins, par rapport à la séquence SEQ ID N°1 , une substitution d'un acide aminé.19. Pharmaceutical composition according to claim 16, characterized in that the ESM-1 protein antagonist compound is a polypeptide comprising at least 10 consecutive amino acids of the ESM-1 protein of sequence SEQ ID No. 1, comprising a sequence d amino acids ranging from amino acid at position 119 to amino acid at position 139 of the sequence SEQ ID No. 1 and comprising at least, with respect to the sequence SEQ ID No. 1, a substitution of an amino acid.
20. Composition pharmaceutique selon la revendication 16, caractérisée en ce que le composé antagoniste de la protéine ESM-1 consiste en un polynucléotide antisens. 20. Pharmaceutical composition according to claim 16, characterized in that the ESM-1 protein antagonist compound consists of an antisense polynucleotide.
EP01993475A 2000-11-09 2001-11-08 Use of a compound antagonist of the esm-1 protein for producing a medicine for treating cancer Withdrawn EP1335745A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
FR0014422A FR2816214B1 (en) 2000-11-09 2000-11-09 USE OF ANTAGONISTIC ESM-1 PROTEIN COMPOUND FOR THE MANUFACTURE OF A MEDICAMENT FOR THE PREVENTION AND / OR TREATMENT OF CANCER
FR0014422 2000-11-09
PCT/FR2001/003475 WO2002038178A1 (en) 2000-11-09 2001-11-08 Use of a compound antagonist of the esm-1 protein for producing a medicine for treating cancer

Publications (1)

Publication Number Publication Date
EP1335745A1 true EP1335745A1 (en) 2003-08-20

Family

ID=8856265

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01993475A Withdrawn EP1335745A1 (en) 2000-11-09 2001-11-08 Use of a compound antagonist of the esm-1 protein for producing a medicine for treating cancer

Country Status (7)

Country Link
US (1) US7306797B2 (en)
EP (1) EP1335745A1 (en)
JP (1) JP4121852B2 (en)
AU (1) AU2002218342A1 (en)
CA (1) CA2429404A1 (en)
FR (1) FR2816214B1 (en)
WO (1) WO2002038178A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006506966A (en) * 2002-07-01 2006-03-02 ファルマシア・コーポレーション ESM-1 gene differentially expressed in angiogenesis, its antagonists and methods of using them
US20070197452A1 (en) * 2006-02-17 2007-08-23 Mclaurin Joanne Treatment of amyloid-related diseases
JP2011502111A (en) * 2007-10-24 2011-01-20 アンスティチュ パストゥール ドゥ リール Use of non-glycanated polypeptides in cancer therapy
TWI772927B (en) * 2015-03-31 2022-08-01 德商英麥提克生物技術股份有限公司 Novel peptides and combination of peptides and scaffolds for use in immunotherapy against renal cell carcinoma (rcc) and other cancers

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5747280A (en) * 1995-06-05 1998-05-05 Human Genome Sciences, Inc. Human vascular IBP-like growth factor
AU710568B2 (en) * 1994-12-09 1999-09-23 Human Genome Sciences, Inc. Human vascular IBP-like growth factor
US6670328B1 (en) 1997-06-24 2003-12-30 Institut Pasteur De Lille Proteins and peptides derived from protein ESM-1 and their uses in the treatment and diagnosis of diseases linked to leukocyte migration
FR2775691B1 (en) * 1998-03-05 2000-12-15 Pasteur Institut SPECIFIC MONOCLONAL ANTIBODIES OF ESM-1 PROTEIN, AND USE OF SUCH ANTIBODIES FOR DETECTION OF ESM-1 PROTEIN

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0238178A1 *

Also Published As

Publication number Publication date
AU2002218342A1 (en) 2002-05-21
US20040234526A1 (en) 2004-11-25
FR2816214B1 (en) 2005-10-21
WO2002038178A1 (en) 2002-05-16
FR2816214A1 (en) 2002-05-10
US7306797B2 (en) 2007-12-11
JP4121852B2 (en) 2008-07-23
CA2429404A1 (en) 2002-05-06
JP2004518638A (en) 2004-06-24

Similar Documents

Publication Publication Date Title
JP4689956B2 (en) Growth factor isoform
EP1814579B1 (en) Mutated netrin-4, fragments thereof and their use as medicines
JP3837494B2 (en) Soluble RAGE protein
JP2002518010A (en) 94 human secreted proteins
JP2002512038A (en) Methods for inhibiting tumor invasion or spread in a subject
JPH11513883A (en) Human vascular endothelial growth factor 2
WO2001005422A2 (en) Use of a polypeptide for detecting, preventing or treating a pathological condition associated with a degenerative, neurological or autoimmune disease
JP2002533058A (en) 97 human secreted proteins
JP2004522742A (en) TREM-1 splice variants for use in modifying an immune response
KR20010086070A (en) Promotion or Inhibition of Angiogenesis and Cardiovascularization
JP2002505873A (en) Vascular endothelial growth factor 2
JP2002508167A (en) 110 human secreted proteins
JP2001514885A (en) 70 human secreted proteins
JP2001514024A (en) 50 human secreted proteins
JP2002537796A (en) Human glycosylation enzyme
WO2006068822A1 (en) Notch receptor agonists and uses
JP2002505871A (en) 31 human secretory proteins
JP2002514925A (en) 19 human secreted proteins
JP4426575B2 (en) Novel endogenous bioactive peptide that suppresses cardiac function / hypertensive
EP1335745A1 (en) Use of a compound antagonist of the esm-1 protein for producing a medicine for treating cancer
DE60038740T2 (en) Stimulation or inhibition of angiogenesis and cardiovascularisation
JPWO2002033094A1 (en) Antibodies that inhibit VPLF activity
JP2003521215A (en) 83 human secreted proteins
CA2558754A1 (en) Glycopeptides derived from pancreatic structures, antibodies and applications thereof in diagnostics and therapeutics
JP4171228B2 (en) Soluble type RAGE measurement method

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030610

AK Designated contracting states

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

RIN1 Information on inventor provided before grant (corrected)

Inventor name: TONNEL, ANDRE-BERNARD

Inventor name: BECHARD, DAVID

Inventor name: LASSALLE, PHILIPPERESIDENCE SEBASTOPOL

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1058758

Country of ref document: HK

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 35/00 20060101ALI20090409BHEP

Ipc: G01N 33/53 20060101ALI20090409BHEP

Ipc: A61K 31/7088 20060101ALI20090409BHEP

Ipc: A61K 38/17 20060101ALI20090409BHEP

Ipc: A61K 39/395 20060101AFI20090409BHEP

GRAC Information related to communication of intention to grant a patent modified

Free format text: ORIGINAL CODE: EPIDOSCIGR1

GRAC Information related to communication of intention to grant a patent modified

Free format text: ORIGINAL CODE: EPIDOSCIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20100615

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1058758

Country of ref document: HK